Note: Descriptions are shown in the official language in which they were submitted.
CA 02732323 2015-12-10
55929-4
TGR5 MODULATORS AND METHODS OF USE THEREOF
Field of the Invention
The invention relates to compounds containing a sulfate or sulfonic acid
moiety that modulate TGR5 and pharmaceutical compositions containing such
compounds
useful in methods for the treatment and prevention of disease.
Background of the Invention
TGR5 receptor is a G-protein-coupled receptor that has been identified as a
cell-
surface receptor that is responsive to bile acids (BAs). The primary structure
of TGR5 and its
responsiveness to bile acids has been found to be highly conserved in TGR5
among human,
bovine, rabbit, rat, and mouse, and thus suggests that TGR5 has important
physiological
functions. TGR5 has been found to be widely distributed in not only lymphoid
tissues but
also in other tissues. High levels of TGR5 mRNA have been detected in
placenta, spleen,
and monocytes/macrophages. Bile acids have been shown to induce
internalization of the
TGR5 fusion protein from the cell membrane to the cytoplasm. ICawamata et al.
2003, J. Bio.
Chem., 278, 9435. TGR5 has been found to be identical to hGPCR19 reported by
Takeda et
al. 2002, FEBS Lett. 520, 97-101.
TGR5 is associated with the intracellular accumulation of cAMP, that is widely
expressed in diverse cell types. While the activation of this membrane
receptor in
macrophages decreases pro-inflammatory cytokine production, (Kawamata, Y.;
Fujfi, R.;
Hosoya, M.; Harada, M.; Yoshida, H.; Miwa, M.; Fukusumi, S.; Habata, Y.; Itoh,
T.;
Shintani, Y.; Hinuma, S.; Fujisawa, Y.; Fujin , M., A G protein-coupled
receptor responsive
to bile acids. J. Biol. Chem. 2003, 278, 9435-9440) the stimulation of TGR5 by
BAs in
adipocytes and myocytes enhances energy expenditure (Watanabe, M.; Houten, S.
M.;
Mataki, C.; Christoffolete, M. A.; Kim, B. W.; Sato, H.; Messaddeq, N.;
Harney, J. W.;
Ezaki, 0.; Kodama, T.; Schoonjans, K..; Bianco, A. C.; Auwerx, J., Bile acids
induce energy
expenditure by promoting intracellular thyroid hormone activation. Nature.
2006, 439,
484-489). This latter effect involves the cAMP-dependent induction of type 2
iodothyronine
deiodinase (D2), which by, locally converting T4 into T3, gives rise to
increased thyroid
hormone activity. Consistent with the role of TGR5 in the control of energy
metabolism,
female TGR5 knock-out mice show a significant fat accumulation with body
weight gain when
challenged with a high fat diet, indicating that the lack of TGR5 decreases
energy expenditure
- 1 -
,
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
and elicits obesity (Maruyama, T.; Tanaka, K.; Suzuki, J.; Miyoshi, H.;
Harada, N.; Nakamura,
T.; Miyamoto, Y.; Kanatani, A.; Tamai, Y., Targeted disruption of G protein-
coupled bile acid
receptor 1 (Gpbarl/MBar) in mice. J. Endocrinol. 2006, 191, 197-205). In
addition and in line
with the involvement of TGR5 in energy homeostasis, bile acid activation of
the membrane
receptor has also been reported to promote the production of glucagon-like
peptide 1 (GLP-1)
in murine enteroendocrine cell lines (Katsuma, S.; Hirasawa, A.; Tsujimoto,
G., Bile acids
promote glucagon-like peptide-1 secretion through TGR5 in a murine
enteroendocrine cell
line STC-1. Biochem. Biophys. Res. Commun. 2005, 329, 386-390). On the basis
of all the
above observations, TGR5 is an attractive target for the treatment of disease.
Few examples of TGR5 agonists have been so far described in literature.
Recently, 23-
alkyl-substituted and 6,23-alkyl-disubstituted derivatives of chenodeoxycholic
acid, such as
the 6a-ethyl-23(S)-methyl-chenodeoxycholic acid shown below, have been
reported as
potent and selective agonists of TGR5 (Pellicciari, R.; Sato, H.; Gioiello,
A.; Costantino, G.;
Macchiarulo, A.; Sadeghpour, B. M.; Giorgi, G.; Schoonjans, K.; Auwerx, J.,
Nongenomic
actions of bile acids. Synthesis and preliminary characterization of 23-and
6,23-alkyl-
substituted bile acid derivatives as selective modulators for the g-protein
coupled receptor
TGR5. J. Med. Chem. 2007, 50, 4265-4268).
-,
23
-
CO2H
. O.
HO"' 6- '''OH
H _
/
In particular, methylation at the C23-(S) position of natural BAs confers a
marked selectivity
to TGR5 over FXR (farnesoid X receptor) activation, whereas the 6a-alkyl
substitution
increases the potency at both receptors. Other TGR5 agonists include 6-methy1-
2-oxo-4-
thiophen-2-y1-1,2,3,4-tetrahydro-pyrimidine-5-carboxylic acid benzyl ester
(W0004067008,
Takeda Chemical Industries LTD, Japan, 2004) and oleanoic acid (Sato, H.;
Genet, C.;
Strehle, A.; Thomas, C.; Lobstein, A.; Wagner, A.; Mioskowski, C.; Auwerx, J.;
Saladin, R.,
Anti-hyperglycemic activity of a TGR5 agonist isolated from Olea europaea.
Biochem.
- 2 -
i
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
and Biophys. Res. Commun. 2007, 362, 793-798; Ito, F.; Hinuma, K.; Kanzald,
N.; Mild, T.;
Kawamata, Y.; 0i, S.; Tawaeaishi, T.; Ishichi, Y.; Hirohashi, M. Preparation
of aromatic ring-
fused cyclic compounds as TGR5 receptor agonists. PN: W02004067008, 2004. More
recently,
the first synthesis of enantiomeric chenodeoxycholic acid (CDCA) and
lithocholic acid
(LCA) has allowed to assess the specificity of the interaction of natural BAs
to TGR5
(Katona, B. W.; Cummins, C. L.; Ferguson, A. D.; Li, T.; Schmidt, D. R.;
Mangelsdorf, D. J.;
Covey, D. F., Synthesis, Characterization, and Receptor Interaction Profiles
of Enantiomeric
Bile Acids. J. Med. Chem. 2007, 50, 6048-6058).
While these chemical tools have provided for the first time a pharmacological
differentiation of genomic versus nongenomic effects of BAs, some of them also
allowed to
draw a first structure-activity relationship study where the presence of an
accessory binding
pocket in TGR5 plays a pivotal role in determining ligand selectivity
(Pellicciaii, R.; Sato,
H.; Gioiello, A.; Costantino, G.; Macchiarulo, A.; Sadeghpour, B. M.; Giorgi,
G.;
Schoonjans, K.; Auwerx, J., Nongenomic actions of bile acids. Synthesis and
preliminary
characterization of 23-and 6,23-alkyl-substituted bile acid derivatives as
selective modulators
for the g-protein coupled receptor TGR5. J. Med. Chem. 2007, 50, 4265-4268).
In this
context, the availability of more potent and selective TGR5 modulators is
necessary to further
identify additional features affecting receptor activation and characterize
the physiological
and pharmacological actions of this receptor.
There is a need for the development of TGR5 modulators for the treatment and
prevention of disease. The present invention has identified compounds, which
contain a
sulfate or sulfonic acid moiety, that modulate TGR5 as well as methods of
using these
compounds to treat disease.
Summary of the Invention
The present invention relates to TGR5 modulators containing a sulfate or
sulfonic
acid moiety and their use to treat and prevent diseases that involve
modulation of the TGR5
receptor, such as metabolic disease, inflammatory disease, liver disease,
autoimmune disease,
cardiac disease, kidney disease, cancer and gastrointestinal disease.
The invention includes a compound having the formula A:
- 3 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
R2 (CHR6)m____(CHR5)ii __ (CH2)0-(0)pS03H
&S.
R11'1.0W.'µR1
R12 H R7
R4
(A)
or a salt, solvate, or hydrate thereof, wherein: R1 is hydrogen, hydroxy,
substituted or
unsubstituted alkyl, or halogen; R2 is hydrogen or a-hydroxy; R4 is hydrogen,
substituted or
unsubstituted alkyl, or halogen; R5 is hydrogen, unsubstituted alkyl, or aryl;
R6 is hydrogen, unsubstituted or substituted alkyl, or R5 and R6 taken
together with the
carbons to which they are attached form a ring of size 3, 4, 5, or 6 atoms; R7
is hydrogen,
substituted or unsubstituted alkyl, or hydroxy; R11 is hydroxyl, OSO3H, 0S03-,
OCOCH3,
OPO3H, 0P032- or hydrogen; R12 is hydroxyl, OSO3H, 0S03", OCOCH3, OPO3H, 0P032-
or
hydrogen, or taken together R11 and R12 form a carbonyl;
m is 0, 1 or 2 ; n is 0 or 1; o is 0, or 1; and p is 0 or 1; provided that
(1) when m + n + o = 3 or 4, p is zero, and R5 is hydrogen, then R4 is not
hydrogen, unless R7
is OH; (2) when m + n + o = 3, p is 1, and R5 and R6 are each hydrogen, then
at least one of
R2 and R4 is not hydrogen; and (3) when m + n + o =2, then at least one of R5
and R6 is not
hydrogen.
In one aspect, the invention includes a compound having the formula D:
R2
03H
1110111
R11'1.55.µ`Ri
R12 Hi R7
R4
(D)
or a salt, solvate, or hydrate thereof, wherein R1 is hydrogen, hydroxy,
substituted or
unsubstituted alkyl, or halogen; R2 is hydrogen or a-hydroxy; RI is hydrogen,
substituted or
unsubstituted alkyl, or halogen; R7 is hydrogen, substituted or unsubstituted
alkyl, or
hydroxy; R11 is hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H, 0P032, or hydrogen; and
R12
- 4 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
is hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H, 0P032-, or hydrogen, or taken
together R11
and R12 form a carbonyl, provided that R4 is not hydrogen, unless R7 is OH.
In one aspect, the invention includes a compound having the formula E:
R6
OSO3H
R2
R5
R11" " Ri
R12 H: R7
R4
(E)
or a salt, solvate, or hydrate conjugate thereof, wherein: R1 is hydrogen,
hydroxy, substituted
or unsubstituted alkyl, or halogen; R2 is hydrogen or a-hydroxyl; R4 is
hydrogen, substituted
or unsubstituted alkyl, or halogen; R5 is hydrogen, unsubstituted alkyl, or
aryl;
R6 is hydrogen, unsubstituted or substituted alkyl, or R5 and R6 taken
together with the
carbons to which they are attached form a ring of size 3, 4, 5, or 6 atoms; R7
is hydrogen,
substituted or unsubstituted alkyl, or hydroxy; R11 is hydroxyl, OSO3H, 0S03-,
OCOCH3,
OPO3H, OP032-, or hydrogen; and R12 is hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H,
0P032-, or hydrogen, or taken together R11 and R12 form a carbonyl, provided
that at least one
of R5 or R6 is not hydrogen.
In one aspect, the invention includes a compound or a salt, solvate, or
hydrate thereof,
wherein R1 is OH. In one aspect, the invention includes a compound or a salt,
solvate, or
hydrate thereof, wherein R7 is H. In one aspect, the invention includes a
compound or a salt,
solvate, or hydrate thereof, wherein R2 is H. In one aspect, the invention
includes a
compound or a salt, solvate, or hydrate thereof, wherein R4 is unsubstituted
alkyl.
In one aspect, the invention includes a compound selected from
- 5 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
OSO3H OH '-
ISM SO3H
'Ha* 'OH Has, 11141.õ
H H- 0
(2A), (3A)
0
0-S-0--H
S 0"
01111410.,,
H OH O3H 2
H
(4A), (8A),
OH
0
,,S
2 to, `0,
HO'IPH
and (9A) or a salt, solvate, or hydrate
thereof.
In one aspect, the invention includes a compound that is a pharmaceutically
acceptable salt. In one aspect, the invention includes a pharmaceutical
composition
comprising a compound or a salt, solvate, or hydrate thereof and at least one
pharmaceutically acceptable excipient.
In another aspect, the invention includes use of a compound having the formula
A or a
salt, solvate, or hydrate thereof, or a pharmaceutical composition comprising
a compound
having the formula A or a salt, solvate, or hydrate thereof:
R2 (CHR6)õ_(CHR5)¨(CH2)0 _________ (0)pS03H
S.
R12 Hi R7
R4
(A)
wherein:
- 6 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
R1 is hydrogen, hydroxy, substituted or unsubstituted alkyl, or halogen; R2 is
hydrogen or a-
hydroxy; R4 is hydrogen, substituted or unsubstituted alkyl, or halogen; R5 is
hydrogen,
unsubstituted alkyl, or aryl; R6 is hydrogen, unsubstituted or substituted
alkyl, or R5 and R6
taken together with the carbons to which they are attached form a ring of size
3, 4, 5, or 6
atoms; R7 is hydrogen, substituted or unsubstituted alkyl, or hydroxy; R is
hydroxyl,
OSO3H, 0S03-, OCOCH3, 0P0311, 0P032" or hydrogen;
R12 is hydroxyl, OSO3H, 0S03", OCOCH3, OPO3H, 0P032, or hydrogen, or taken
together
Rii and R12 form a carbonyl;
m is 0, 1, or 2; n is 0 or 1; o is 0 or 1; and p is 0 or 1, in the manufacture
of a medicament for
a treating or preventing a disease in a subject that involves modulation of
the TGR5 receptor.
In one aspect, the invention includes the use, wherein the disease is selected
from
metabolic disease, inflammatory disease, liver disease, autoinunune disease,
cardiac disease,
kidney disease, cancer, and gastrointestinal disease. In one aspect, the
invention includes the
use, wherein the disease is selected from inflammatory disease and cancer. In
one aspect, the
invention includes the use, wherein the compound or pharmaceutical composition
is
administered to the subject orally, parentally, intravenously, or topically.
In one aspect, the
invention includes the use, wherein the subject is a human.
The above description sets forth rather broadly the more important features of
the
present invention in order that the detailed description thereof that follows
may be
understood, and in order that the present contributions to the art may be
better appreciated.
Other objects and features of the present invention will become apparent from
the following
detailed description considered in conjunction with the examples.
Brief Description of the Figures
Figure 1 is a graph that depicts the surface tension plotted against the
logarithm of the
concentration of 3A (mM) in NaC1 0.15M.
Figure 2 is a graph that depicts that metabolic stability of 2A in simulated
pancreatic
fluid.
Figure 3 is a bile flow chart for a duodenal infusion experiment performed
using 3A.
Figure 4 is a bile flow chart for a femoral infusion experiment performed
using 3A.
- 7 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Figure 5 is a graph that depicts secretion rates verses time in femoral and
duodenal
infusion experiments performed using 3A.
Figures 6 and 7 are graphs that shows 3A and its main metabolite identified in
bile
using mass spectrometry in a duodenal infusion experiment. Data are reported
as absolute
area values. Figure 7 is a zoom display of Figure 6.
Figure 8 is a graph that shows the evolution of weigh gain after treatment (%)
(Example 11).
Figure 9 is a series of bar graphs the evolution of body composition after
treatment
(%) 5 weeks after treatment (Example 11).
Figure 10 shows an assessment of glucose homeostasis for compound 3A. Figure
10A shows glycemia 3 weeks after treatment. Figure 10B shows fructosamines 3
weeks after
treatment. Figure 10C shows the results of a glucose tolerance test 9 weeks
after treatment.
Description of the Invention
The details of one or more embodiments of the invention are set forth in the
accompanying description below. Although any methods and materials similar or
equivalent
to those described herein can be used in the practice or testing of the
present invention, the
methods and materials are now described. Other features, objects, and
advantages of the
invention will be apparent from the description. In the specification, the
singular forms also
include the plural unless the context clearly dictates otherwise. Unless
defined otherwise, all
technical and scientific terms used herein have the same meaning as commonly
understood
by one of ordinary skill in the art to which this invention belongs. In the
case of conflict, the
present specification will control.
In one aspect, the invention provides a compound having the formula A:
R2 (CH R6)m¨(CHROn ____ (CH2)0¨(0)pS03H
S.
R12 H:F.; R7
(A) or
a salt, solvate, or hydrate, wherein R1 is hydrogen, hydroxy, substituted or
unsubstituted
alkyl, or halogen; R2 is hydrogen or a-hydroxy; R4 is hydrogen, substituted or
unsubstituted
alkyl, or halogen; R5 is hydrogen, unsubstituted alkyl, or aryl; R6 is
hydrogen, unsubstituted
- 8 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
or substituted alkyl, or R5 and R6 taken together with the carbons to which
they are attached
form a ring of size 3, 4, 5, or 6 atoms; R7 is hydrogen, substituted or
unsubstituted alkyl, or
hydroxy; RH is hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H, 0P032- or hydrogen; R12
is
hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H, 0P032- or hydrogen, or taken together
Rii and
R12 form a carbonyl; m is 0, 1 or 2; n is 0 or 1; o is 0 or 1; and p is 0 or
1.
In one aspect, the invention provides that when m + n + o =3 or 4, p is zero,
and R5 is
hydrogen, then R4 is not hydrogen, unless R7 is OH. In another aspect, the
invention provides
that when m + n + o = 3 or 4, p is zero, and R5 is not unsubstituted alkyl or
aryl, then R4 is
not hydrogen, unless R7 is OH.
In one aspect, the invention provides that when m + n + o = 3, p is 1, and R5
and R6
are each hydrogen, then at least one of R2 and R4 is not hydrogen.
In one aspect, the invention provides that when m + n + o =2, and at least one
of R5
and R6 is not hydrogen.
In one aspect, the invention does not include compounds A, B, and C:
ee OSO3H
SO3H
.1101110.,
HO's 'OH HO" 'OH
H (A), H (B), and
OSO3H
= 410.:1111
H 'OH
(C) or a salt, solvate, or hydrate thereof. In another
aspect, the invention does not include compound 6A or a salt, solvate, or
hydrate thereof. In
another aspect, the invention does not include compound 7A or a salt, solvate,
or hydrate
thereof.
In one aspect, the invention provides a compound having the formula B:
- 9 -
_
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
..,
R2 -- (CHR6),õ_(CHR5)p-(CH2)0-(0)pS03H
HO"' '''0H
H m=
rm
(B) or
a salt, solvate, or hydrate thereof, wherein R2, R4, R5, and R6, III, n, o,
and p are as described
above. In one aspect, the invention provides that when m + n + o = 3 or 4, p
is zero, R5 is
hydrogen, then R4 is not hydrogen. In one aspect, the invention provides that
when m + n + o
= 3, p is 1, and R5 and R6 are each hydrogen, then at least one of R2 and R4
is not hydrogen.
_
In another aspect, the invention provides that when m + n + o =2, then at
least one of R5 and
R6 is not hydrogen.
In one aspect, the invention provides a compound having the formula C:
R2 ''= (CHR6)m(CHR5)p-(CH2)0 ______________________________ (0)pS03H
HO" 'ccs
_ '''OH -
H =
(C) or a salt, solvate, or hydrate thereof, wherein R2, R5, R6, Ill, n, o, and
p are as described
above. In one aspect, when m + n + o = 2, then at least one of R5 and R6 is
not hydrogen.
In one aspect, the invention provides a compound having the formula D:
,
R2 -,
03H
Os
R11/1,
R12 H;, R7
rµzi
(D) or a salt, solvate, or hydrate
thereof, wherein R1, R2, R4, R7, Rli, and R12 are as described above. In one
aspect, R4 is not
hydrogen.
- 10 -
,
i
i
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
In one aspect, the invention provides a compound having the formula E:
R6
IR2
OSO3H
. R5
R111". OWp"µR1
R12 H R7
R4
(E) or a salt, solvate, or hydrate
thereof, wherein: R1 is hydrogen, hydroxy, substituted or unsubstituted alkyl,
or halogen; R2
is hydrogen or a-hydroxyl; R4 is hydrogen, substituted or unsubstituted alkyl,
or halogen;
R5 is hydrogen, unsubstituted alkyl, or aryl; R6 is hydrogen, unsubstituted or
substituted alkyl,
or R5 and R6 taken together with the carbons to which they are attached form a
ring of size 3,
4, 5, or 6 atoms; R7 is hydrogen, substituted or unsubstituted alkyl, or
hydroxy; Rii is
hydroxyl, OSO3H, 0S03-, OCOCH3, OPO3H, 0P032- or hydrogen; and R12 is
hydroxyl,
OSO3H, 0S03-, OCOCH3, OPO3H, 0P032- or hydrogen, or taken together R and R12
form a
carbonyl. In one aspect, the invention provides that at least one of R5 or R6
is not hydrogen.
In one aspect, the invention provides a compound having the formula F:
R2
OSO3H
10011 R5
R11'1.
R12
R1
R12 H R7
R4
(F) or a salt, solvate, or hydrate
thereof, wherein R5 is unsubstituted or substituted alkyl or aryl; and
R1, R2, R4, R7, R11, and R12 are as described above.
In one aspect, the invention provides a compound having the formula G:
- 11 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
R2
OSO3H
S.
R11' '
R12 H R7
R4
(G) or a salt, solvate, or
hydrate thereof, wherein RI, R2, R4, R7, R11, and R12 are as described above.
In one aspect, the invention provides a compound having the formula H:
R6
OSO3H
R2
OS. R5
H z
(H) or a salt, solvate, or hydrate thereof,
wherein R2, R5 and R6 are as described above.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R1 is OH.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R7 is H.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R1 is OH and R7 is H.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R2 is H. In one aspect, the invention provides a or a salt, solvate,
or hydrate thereof,
wherein R2 is alpha-OH. In one aspect, the invention provides a salt, solvate,
or hydrate
thereof, wherein R2 is beta-OH.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R4 is unsubstituted alkyl. In one aspect, the invention provides a
compound or a salt,
solvate, or hydrate thereof, wherein R4 is ethyl. In one aspect, the invention
provides a
compound or a salt, solvate, or hydrate thereof, wherein R4 is methyl.
-12-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R5 is not hydrogen. In one aspect, the invention provides a compound
or a salt,
solvate, or hydrate thereof, wherein R5 is unsubstituted alkyl. In one aspect,
the invention
provides a compound or a salt, solvate, or hydrate thereof, wherein R5 is
methyl.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R6 is hydrogen.
In one aspect, the invention provides a compound of salt, solvate, or hydrate
thereof,
wherein R5 and Ro are both hydrogen.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R is hydroxyl. In one aspect, the invention provides a compound or a
salt, solvate,
or hydrate thereof, wherein R12 is hydrogen. In one aspect, the invention
provides a
compound or a salt, solvate, or hydrate thereof, wherein R11 is hydroxyl and
R12 is hydrogen.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein m is 1. In one aspect, the invention provides a compound or a salt,
solvate, or
hydrate thereof, wherein m is 2. In one aspect, the invention provides a
compound or a salt,
solvate, or hydrate thereof, wherein n is 1.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein o is 0. In one aspect, the invention provides a compound or a salt,
solvate, or hydrate
thereof, wherein o is 1.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein p is 1. In one aspect, the invention provides a compound or a salt,
solvate, or hydrate
thereof, wherein p is 0.
In one aspect, the invention provides a compound or a salt, solvate, or
hydrate thereof,
wherein R4 is ethyl and p is 1. In one aspect, the invention provides a
compound or a salt,
solvate, or hydrate thereof, wherein R4 is ethyl and the compound contains a
sulfonate
moiety. In one aspect, the invention provides a compound or a salt, solvate,
or hydrate
thereof, wherein R4 is hydrogen or substituted or unsubstituted alkyl and p is
0.
In one aspect, the invention provides a compound selected from
- 13
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
=
OSO3H OH
SO3H
Has. 111Nr_ = ''OH =H047 OH
H z
OH
SO3H ee SO3H
Ilk
HO" 'OH
0
OH
0¨S-0¨H
101) SO3H
2 8
HO"'OH
010
HO" OH H z
OH
el* 20/ %
Has." '''0H
and or a salt, solvate, or hydrate thereof.
In one aspect, the invention provides a compound, wherein the compound is a
pharmaceutically acceptable salt.
In one aspect, the invention provides a pharmaceutically acceptable salt
selected
0S03-N a+ OH --
1011111 03-NI a+
s.1141. '0111H HO`µ.
H
from Ha
- 14 -
,
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
OH '-
O. 03-Na+ O. 03-Na+
'Has.H.1z. 'OH
''OH
0
OH 0¨S-0-Na+
&O. S03-Na+
HO"
0
'Ha* 'OH
2
OH
and
OH
,0
O.
0/ 0-NaHO"+
.1110õ
. 'OH
H
In one aspect, the invention provides a pharmaceutical composition comprising
a
compound and at least one pharmaceutically acceptable excipient.
In one aspect, the invention provides the use of a compound or a
pharmaceutical
composition of the invention, in the manufacture of a medicament for treating
or preventing a
disease in a subject. In another aspect, the invention provides a method of
treating or
preventing disease in a subject by administering a compound or a
pharmaceutical
composition of the invention. In one aspect, the invention provides a
therapeutically
effective amount of a compound or pharmaceutical composition of the invention
is
administered to the subject. In one aspect, the invention provides a
prophylactially effective
amount of a compound or pharmaceutical composition of the invention is
administered.
In one aspect, the invention provides the use of the compound or
pharmaceutical
composition of the invention, in the manufacture of a medicament for a
treating or preventing
a disease in a subject that involves modulation of the TGR5 receptor. The
invention includes
a method of treating or preventing a disease that involves modulation of the
TGR5 receptor in
a subject by administering a compound or pharmaceutical composition of the
invention.
- 15 -
_ _ ,
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
In one aspect, the invention provides the use, wherein the disease is selected
from
metabolic disease, inflammatory disease, liver disease, autoimmune disease,
cardiac disease,
kidney disease, cancer, and gastrointestinal disease using a compound having
the formula A,
B, C, D, E, F, or G. The invention includes a method of treating or preventing
a disease
selected from metabolic disease, inflammatory disease, liver disease,
autoimmune disease,
cardiac disease, kidney disease, cancer, and gastrointestinal disease using a
compound of
formula A, B, C, D, E, F, or G. The invention includes a method of treating or
preventing
inflammatory disease or cancer using a compound of formula A, B, C, D, E, F,
or G.
In one aspect, the invention provides for the use, wherein the disease is a
metabolic
disease selected from obesity, diabetes, metabolic syndrome, insulin
resistance, hypertension,
and dyslipidemia. The invention includes a method of treating or preventing a
metabolic
disease selected from obesity, diabetes, metabolic syndrome, insulin
resistance, hypertension,
and dyslipidemia.
In one aspect, the invention provides for the use, wherein the disease is an
inflammatory disease selected from allergy, osteoarthritis, appendicitis,
bronchial asthma,
pancreatitis, allergic rash, and psoriasis. The invention includes a method of
treating or
preventing an inflammatory disease selected from allergy, osteoarthritis,
appendicitis,
bronchial asthma, pancreatitis, allergic rash, and psoriasis.
In one aspect, the invention provides for the use, wherein the disease is an
autoimmune disease selected from rheumatoid arthritis, multiple sclerosis, and
type I
diabetes. The invention includes a method of treating or preventing an
autoimmune disease
selected from rheumatoid arthritis, multiple sclerosis, and type I diabetes.
In one aspect, the invention provides for the use, wherein the disease is a
gastrointestinal disease selected from inflammatory bowel disease (Crohn's
disease,
ulcerative colitis), short bowel syndrome (post-radiation colitis),
microscopic colitis, irritable
bowel syndrome (malabsorption), and bacterial overgrowth. The invention
includes a
method of treating or preventing a gastrointestinal disease selected from
inflammatory bowel
disease (Crohn's disease, ulcerative colitis), short bowel syndrome (post-
radiation colitis),
microscopic colitis, irritable bowel syndrome (malabsorption), and bacterial
overgrowth.
In one aspect, the invention provides for the use, wherein the disease is
kidney disease
selected from diabetic nephropathy, chronic renal failure, hypertensive
nephrosclerosis,
chronic glomerulonephritis, chronic transplant glomerulopathy, chronic
interstitial nephritis,
and polysystic kidney disease. The invention includes a method of treating or
preventing
-16-
_ ,
,
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
kidney disease selected from diabetic nephropathy, chronic renal failure,
hypertensive
nephrosclerosis, chronic glomerulonephritis, chronic transplant
glomerulopathy, chronic
interstitial nephritis, and polysystic kidney disease.
In one aspect, the invention provides for the use, wherein the disease is
cancer
selected from colorectal cancer, liver cancer, heptacellular carcinoma,
cholangio carcinoma,
renal cancer, gastric cancer, pancreatic cancer, prostate cancer, and
insulanoma. The
invention includes a method of treating or preventing cancer selected from
colorectal cancer,
liver cancer, heptacellular carcinoma, cholangio carcinoma, renal cancer,
gastric cancer,
pancreatic cancer, prostate cancer, and insulanoma.
In one aspect, the invention provides for the use, wherein the disease is a
liver disease
selected from nonalcoholic steatohepatitis, nonalcoholic fatty liver disease,
chronic viral
hepatitis, alcoholic liver disease, drug induced hepatitis, hemochromatosis,
primary biliary
cirrhosis, primary sclerosing cholangitis, portal hypertension, bile
desaturation, Gaucher's
disease, Wilson's disease, al -antitrypsin deficiency, total parenteral
nutrition (TPN),
cholelithiasis, TPN-associated cholestasis and sepsis. The invention includes
a method of
treating or preventing a liver disease selected from nonalcoholic
steatohepatitis, nonalcoholic
fatty liver disease, chronic viral hepatitis, alcoholic liver disease, drug
induced hepatitis,
hemochromatosis, primary biliary cirrhosis, primary sclerosing cholangitis,
portal
hypertension, bile desaturation, Gaucher's disease, Wilson's disease, a 1 -
antitrypsin
deficiency, total parenteral nutrition (TPN), cholelithiasis, TPN-associated
cholestasis and
sepsis.
In one aspect, the invention provides for the use, wherein the cardiac disease
is
selected from congestive heart failure, myocardial infarction,
atherosclerosis, angina pectoris,
arteriosclerosis and cerebrovascular disease (hemorrhage, stroke,
cerebrovascular infarction).
The invention includes a method of treating or preventing a cardiac disease
selected from
congestive heart failure, myocardial infarction, atherosclerosis, angina
pectoris,
arteriosclerosis and cerebrovascular disease (hemorrhage, stroke,
cerebrovascular infarction).
In one aspect, the disease involves modulation of the TGR5 receptor. In one
aspect,
the compound is a TGR5 agonist. In one aspect, the compound is a selective
TGR5 agonist
over FXR activator. In one aspect, the compound is a partial modulator of FXR.
In one
aspect, the compound is a partial FXR agonist.
- 17 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
In one aspect, the compound or pharmaceutical composition of the invention is
administered to the subject orally, parentally, intravenously, or topically.
In one aspect, the
subject is a human.
Definitions
For convenience, certain terms used in the specification, examples and
appended
claims are collected here.
The term "treating", as used herein, means relieving, lessening, reducing,
eliminating,
modulating, or ameliorating, i.e. causing regression of the disease state or
condition. Treating
can also include inhibiting, i.e. arresting the development, of a existing
disease state or
condition, and relieving or ameliorating, i.e. causing regression of an
existing disease state or
condition, for example when the disease state or condition may already be
present.
The term "preventing", as used herein means, to completely or almost
completely stop
a disease state or condition, from occurring in a patient or subject,
especially when the patient
or subject is predisposed to such or at risk of contracting a disease state or
condition.
"Alkyl" includes saturated aliphatic groups, including straight-chain alkyl
groups
(e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl,
decyl), branched-chain
alkyl groups (e.g., isopropyl, tert-butyl, isobutyl). In certain embodiments,
a straight chain or
branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., C1-
C6 for straight
chain, C3-C6 for branched chain). In some examples, a straight chain or
branched chain alkyl
has four or fewer carbon atoms in its backbone.
As used herein, "cycloalkyl" is intended to include saturated ring groups,
such as
cyclopropyl, cyclobutyl, or cyclopentyl. C3-8 cycloalkyl is intended to
include C3, C4, C5,
C6, C7, and C8 cycloalkyl groups.
The term "substituted alkyl" refers to an alkyl moieties having a substituent
replace
one or more hydrogen atoms on at least one or more carbons of the hydrocarbon
backbone.
Such substituents can include, for example, alkyl, alkenyl, alkynyl, halogen,
hydroxyl,
alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy,
carboxylate,
alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl,
alkylaminocarbonyl,
dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato,
phosphinato,
cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and
alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino,
carbamoyl
and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate,
sulfates,
-18-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl,
cyano, azido,
heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.
"Aryl" includes groups with aromaticity, including 5- and 6-membered
"unconjugated", or single-ring, aromatic groups that may include from zero to
four
heteroatoms, as well as "conjugated", or multicyclic, systems with at least
one aromatic ring.
Examples of aryl groups include benzene, phenyl, pyrrole, furan, thiophene,
thiazole,
isothiazole, imidazole, triazole, tetrazole, pyrazole, oxazole, isooxazole,
pyridine, pyrazine,
pyridazine, and pyrimidine, and the like. Furthermore, the term "aryl"
includes multicyclic
aryl groups, e.g., tricyclic, bicyclic, e.g., naphthalene, benzoxazole,
benzodioxazole,
benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl,
quinoline,
isoquinoline, napthridine, indole, benzofuran, purine, benzofuran,
deazapurine, or indolizine.
Those aryl groups having heteroatoms in the ring structure may also be
referred to as "aryl
heterocycles", "heterocycles," "heteroaryls" or "heteroaromatics". The
aromatic ring can be
substituted at least one ring position with such substituents as described
above, as for
example, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy,
alkoxycarbonyloxy,
aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminocarbonyl,
aralkylaminocarbonyl,
alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, arallcylcarbonyl,
alkenylcarbonyl,
alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato,
phosphinato,
cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and
alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino,
carbamoyl
and ureido), amidino, imino, sulthydryl, alkylthio, arylthio, thiocarboxylate,
sulfates,
alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl,
cyano, azido,
heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Aryl groups
can also be
fused or bridged with alicyclic or heterocyclic rings, which are not aromatic
so as to form a
multicyclic system (e.g., tetralin, methylenedioxyphenyl).
Unless the number of carbons is otherwise specified, "lower alkyl" includes an
alkyl
group, as defined above, but having from one to ten, for example, from one to
six, carbon
atoms in its backbone structure.
The term "ester" includes compounds and moieties which contain a carbon or a
heteroatom bound to an oxygen atom which is bonded to the carbon of a carbonyl
group. The
term "ester" includes alkoxycarboxy groups such as methoxycarbonyl,
ethoxycarbonyl,
propoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, etc. The alkyl, alkenyi, or
alkynyl
groups are as defined above.
-19-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
The term "hydroxy" or "hydroxyl" includes groups with an -OH or
The term "halogen" includes fluorine, bromine, chlorine, iodine, etc. The term
"perhalogenated" generally refers to a moiety wherein all hydrogens are
replaced by halogen
atoms.
An "anionic group," as used herein, refers to a group that is negatively
charged at
physiological pH. Anionic groups include carboxylate, sulfate, sulfonate,
sulfinate, sulfamate,
tetrazolyl, phosphate, phosphonate, phosphinate, or phosphorothioate or
functional
equivalents thereof. "Functional equivalents" of anionic groups are intended
to include
bioisosteres, e.g., bioisosteres of a carboxylate group. Bioisosteres
encompass both classical
bioisosteric equivalents and non-classical bioisosteric equivalents. Classical
and non-classical
bioisosteres are known in the art (see, e.g., Silverman, R. B. The Organic
Chemistry of Drug
Design and Drug Action, Academic Press, Inc.: San Diego, Calif., 1992, pp.19-
23). Another
anionic group is a carboxylate.
The term "unstable functionality" refers to a substitution pattern that
contains a labile
linkage, e.g., a functionality or bond that is susceptible to hydrolysis or
cleavage under
physiological conditions (e.g., aqueous solutions in the neutral pH range).
Examples of
unstable functionalities include acetals and ketals.
Additionally, the compounds of the present invention, for example, the salts
of the
compounds, can exist in either hydrated or unhydrated (the anhydrous) form or
as solvates
with other solvent molecules. Nonlimiting examples of hydrates include
monohydrates,
dihydrates, etc. Nonlimiting examples of solvates include ethanol solvates,
acetone solvates,
etc.
"Solvates" means solvent addition forms that contain either stoichiometric or
non
stoichiometric amounts of solvent. Some compounds have a tendency to trap a
fixed molar
ratio of solvent molecules in the crystalline solid state, thus forming a
solvate. If the solvent
is water the solvate formed is a hydrate, when the solvent is alcohol, the
solvate formed is an
alcoholate. Hydrates are formed by the combination of one or more molecules of
water with
one of the substances in which the water retains its molecular state as H2O,
such combination
being able to form one or more hydrate.
It will be noted that the structure of some of the compounds of the invention
include
asymmetric carbon atoms. It is to be understood accordingly that the isomers
arising from
such asymmetry (e.g., all enantiomers and diastereomers) are included within
the scope of the
invention, unless indicated otherwise. Such isomers can be obtained in
substantially pure
- 20 -
-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
form by classical separation techniques and by stereochemically controlled
synthesis.
Enantiomers (R- and S-configurations) are named according to the system
developed by R.S.
Calm, C. Ingold, and V. Prelog.
Further, the structures and other compounds discussed in this application
include all
atropic isomers thereof. Atropic isomers are a type of stereoisomer in which
the atoms of
two isomers are arranged differently in space. Atropic isomers owe their
existence to a
restricted rotation caused by hindrance of rotation of large groups about a
central bond. Such
atropic isomers typically exist as a mixture, however as a result of recent
advances in
chromatography techniques, it has been possible to separate mixtures of two
atropic isomers
in select cases.
"Stable compound" and "stable structure" are meant to indicate a compound that
is
sufficiently robust to survive isolation to a useful degree of purity from a
reaction mixture,
and formulation into an efficacious therapeutic agent.
As used herein, the term "analog" refers to a chemical compound that is
structurally
similar to another but differs slightly in composition (as in the replacement
of one atom by an
atom of a different element or in the presence of a particular functional
group, or the
replacement of one functional group by another functional group). Thus, an
analog is a
compound that is similar to or comparable in function and appearance to the
reference
compound.
As defined herein, the term "derivative", e.g., in the term "bile acid
derivatives",
refers to compounds that have a common core 4-membered ring structure, and are
substituted
with various groups as described herein.
The chemical compounds described herein can have asymmetric centers. Compounds
of the present invention containing an asymmetrically substituted atom can be
isolated in
optically active or racemic forms. It is well known in the art how to prepare
optically active
forms, such as by resolution of racemic forms or by synthesis from optically
active starting
materials. Many geometric isomers of olefins, C=N double bonds, and the like
can also be
present in the compounds described herein, and all such stable isomers are
contemplated in
the present invention. Cis and trans geometric isomers of the compounds of the
present
invention are described and can be isolated as a mixture of isomers or as
separated isomeric
forms. All chiral, diastereomeric, racemic, and geometric isomeric forms of a
structure are
intended, unless the specific stereochemistry or isomeric form is specifically
indicated. All
processes used to prepare compounds of the present invention and intermediates
made therein
- 21 -
_
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
are, where appropriate, considered to be part of the present invention. All
tautomers of
shown or described compounds are also, where appropriate, considered to be
part of the
present invention.
The term "bioisostere" refers to a compound resulting from the exchange of an
atom
or of a group of atoms with another, broadly similar, atom or group of atoms.
The
bioisosteric replacement may be physicochemically or topologically based.
Examples of
carboxylic acid bioisosteres include acyl sulfonimides, tetrazoles,
sulfonates, and
phosphonates. See, e.g., Patani and LaVoie, Chem. Rev. 96, 3147-3176 (1996).
The phrase "pharmaceutically acceptable" is art-recognized. In certain
aspects, the
term includes compositions, polymers and other materials and/or dosage forms
which are,
within the scope of sound medical judgment, suitable for use in contact with
the tissues of
human beings and animals without excessive toxicity, irritation, allergic
response, or other
problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically acceptable carrier" is art-recognized, and
includes, for
example, pharmaceutically acceptable materials, compositions or vehicles, such
as a liquid or
solid filler, diluent, excipient, solvent or encapsulating material, involved
in carrying or
transporting any subject composition from one organ, or portion of the body,
to another organ,
or portion of the body. Each carrier must be "acceptable" in the sense of
being compatible
with the other ingredients of a subject composition and not injurious to the
patient. In certain
embodiments, a pharmaceutically acceptable carrier is non-pyrogenic. Some
examples of
materials which may serve as pharmaceutically acceptable carriers include: (1)
sugars, such as
lactose, glucose and sucrose; (2) starches, such as corn starch and potato
starch; (3) cellulose,
and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose
and cellulose
acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8)
excipients, such as cocoa
butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil,
sunflower oil,
sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as
propylene glycol; (11)
polyols, such as glycerin, sorbitol, mamitol and polyethylene glycol; (12)
esters, such as ethyl
oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium
hydroxide and
aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic
saline; (18)
Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and
(21) other non-toxic
compatible substances employed in pharmaceutical formulations.
- 22 -
,
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
"Combination therapy" (or "co-therapy") includes the administration of a
compound
of the invention and at least a second agent as part of a specific treatment
regimen intended to
provide the beneficial effect from the co-action of these therapeutic agents
(i.e., the
compound of the invention and at least a second agent). The beneficial effect
of the
combination includes, but is not limited to, phannacokinetic or
phannacodynamic co-action
resulting from the combination of therapeutic agents. Administration of these
therapeutic
agents in combination typically is carried out over a defined time period
(usually minutes,
hours, days or weeks depending upon the combination selected). "Combination
therapy"
may, but generally is not, intended to encompass the administration of two or
more of these
therapeutic agents as part of separate monotherapy regimens that incidentally
and arbitrarily
result in the combinations of the present invention. "Combination therapy" is
intended to
embrace administration of these therapeutic agents in a sequential manner,
that is, wherein
each therapeutic agent is administered at a different time, as well as
administration of these
therapeutic agents, or at least two of the therapeutic agents, in a
substantially simultaneous
manner. Substantially simultaneous administration can be accomplished, for
example, by
administering to the subject a single capsule having a fixed ratio of each
therapeutic agent or
in multiple, single capsules for each of the therapeutic agents. Sequential or
substantially
simultaneous administration of each therapeutic agent can be effected by any
appropriate
route including, but not limited to, oral routes, intravenous routes,
intramuscular routes, and
direct absorption through mucous membrane tissues. The therapeutic agents can
be
administered by the same route or by different routes. For example, a first
therapeutic agent
of the combination selected may be administered by intravenous injection while
the other
therapeutic agents of the combination may be administered orally.
Alternatively, for
example, all therapeutic agents may be administered orally or all therapeutic
agents may be
administered by intravenous injection. The sequence in which the therapeutic
agents are
administered is not narrowly critical.
"Combination therapy" also embraces the administration of the therapeutic
agents as
described above in further combination with other biologically active
ingredients and non-
drug therapies (e.g., surgery or mechanical treatments) . Where the
combination therapy
further comprises a non-drug treatment, the non-drug treatment may be
conducted at any
suitable time so long as a beneficial effect from the co-action of the
combination of the
therapeutic agents and non-drug treatment is achieved. For example, in
appropriate cases, the
- 23
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
beneficial effect is still achieved when the non-drug treatment is temporally
removed from
the administration of the therapeutic agents, perhaps by days or even weeks.
The terms "parenteral administration" and "administered parenterally" as used
herein
refer to modes of administration other than enteral and topical
administration, usually by
injection, and includes, without limitation, intravenous, intramuscular, intra-
arterial,
intrathecal, intracapsular, intraorbital, intracardiac, intradermal,
intraperitoneal, transtracheal,
subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid,
intraspinal and
intrasternal injection and infusion.
A "therapeutically effective amount" of a compound of the invention, or a
combination of compounds is an amount (quantity or concentration) of compound
or
compounds. In one embodiment, when a therapeutically effective amount of a
compound is
administered to a subject in need of treatment symptoms arising from the
disease are
ameliorated immediately or after administration of the compound one or more
times. The
amount of the compound to be administered to a subject will depend on the
particular
disorder, the mode of administration, co-administered compounds, if any, and
the
characteristics of the subject, such as general health, other diseases, age,
sex, genotype, body
weight and tolerance to drugs. The skilled artisan will be able to determine
appropriate
dosages depending on these and other factors.
The term "prophylactically effective amount" means an amount (quantity or
concentration) of a compound of the present invention, or a combination of
compounds, that
is administered to prevent or reduce the risk of a disease ¨ in other words,
an amount needed
to provide a preventative or prophylactic effect. The amount of the present
compound to be
administered to a subject will depend on the particular disorder, the mode of
administration,
co-administered compounds, if any, and the characteristics of the subject,
such as general
health, other diseases, age, sex, genotype, body weight and tolerance to
drugs.
The term "reducing the risk of', as used herein, means to lower the likelihood
or
probability of a disease from occurring in a patient, especially when the
patient or subject is
predisposed to such occurrence.
A "salt" of a compound of the invention is a product of the compound that
contains an
ionic bond and its typically produced by reacting the compound with either an
acid or a base.
A "pharmaceutically acceptable salt" is a salt suitable for administering to a
subject.
As used herein, a "pharmaceutically acceptable salt" refer to a derivative of
a
disclosed compound wherein the parent compound is modified by making acid or
base salts
-24 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
thereof. Examples of pharmaceutically acceptable salts include, but are not
limited to,
mineral or organic acid salts of basic residues such as amines; alkali or
organic salts of acidic
residues such as carboxylic acids; and the like. The pharmaceutically
acceptable salts include
the conventional non-toxic salts or the quaternary ammonium salts of the
parent compound
formed, for example, from non-toxic inorganic or organic acids. For example,
such
conventional non-toxic salts include, but are not limited to, those derived
from inorganic and
organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic,
acetic, ascorbic,
benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edefic, ethane
disulfonic, ethane
sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic,
glycollyarsanilic,
hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodide,
hydroxymaleic,
hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic,
malic, mandelic,
methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic,
phosphoric,
polygalacturonic, propionic, salicylic, stearic, subacetic, succinic,
sulfamic, sulfanilic,
sulfuric, tannic, tartaric, and toluene sulfonic.
The pharmaceutically acceptable salts of the present invention can be
synthesized
from the parent compound that contains a basic or acidic moiety by
conventional chemical
methods. Generally, such salts can be prepared by reacting the free acid or
base forms of
these compounds with a stoichiometric amount of the appropriate base or acid
in water or in
an organic solvent, or in a mixture of the two; generally, non-aqueous media
like ether, ethyl
acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of
suitable salts are found in
Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company,
Easton, PA,
USA, p. 1445 (1990).
A "pharmaceutical composition" is a formulation containing a compound of the
invention in a form suitable for administration to a subject. In another
embodiment, the
pharmaceutical composition is in bulk or in unit dosage form. The unit dosage
form is any of
a variety of forms, including, for example, a capsule, an IV bag, a tablet, a
single pump on an
aerosol inhaler, or a vial. The quantity of active ingredient (e.g., a
formulation of a
compound of the invention or salts thereof) in a unit dose of pharmaceutical
composition is
an effective amount and is varied according to the particular treatment
involved. One skilled
in the art will appreciate that it is sometimes necessary to make routine
variations to the
dosage depending on the age and condition of the patient. The dosage will also
depend on
the route of administration. A variety of routes are contemplated, including
oral, pulmonary,
rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular,
intraperitoneal,
-25-
/
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
intranasal, and the like. Dosage forms for the topical or transdermal
administration of a
compound of this invention include powders, sprays, ointments, pastes, creams,
lotions, gels,
solutions, patches and inhalants. In another embodiment, the active compound
is mixed
under sterile conditions with a pharmaceutically acceptable carrier, and with
any
preservatives, buffers, or propellants that are required.
The term "flash dose" refers to compound formulations that are rapidly
dispersing
dosage forms.
The term "immediate release" is defined as a release of compound from a dosage
form
in a relatively brief period of time, generally up to about 60 minutes. The
term "modified
release" is defined to include delayed release, extended release, and pulsed
release. The term
"pulsed release" is defined as a series of releases of drug from a dosage
form. The term
"sustained release" or "extended release" is defined as continuous release of
a compound
from a dosage form over a prolonged period.
A "subject" includes mammals, e.g., humans, companion animals (e.g., dogs,
cats,
birds, and the like), farm animals (e.g., cows, sheep, pigs, horses, fowl, and
the like) and
laboratory animals (e.g., rats, mice, guinea pigs, birds, and the like).
Typically, the subject
is human.
Compounds of the invention also include prodrugs or physiologically equivalent
derivatives. A "prodrug" or "physiologically equivalent derivative" includes a
precursor
form of the drug which is metabolically converted in vivo to produce the
active drug. The
invention further contemplates the use of prodrugs which are converted in vivo
to the TGR5
modulating compounds used in the methods of the invention (see, e.g., R. B.
Silverman,
1992, "The Organic Chemistry of Drug Design and Drug Action", Academic Press,
Chp. 8).
Such prodrugs can be used to alter the biodistribution (e.g., to allow
compounds which would
not typically cross the blood-brain barrier to cross the blood-brain barrier)
or the
pharmacokinetics of the TGR5 modulating compound. For example, an anionic
group, e.g., a
carboxylate, sulfate or sulfonate, can be esterified, e.g., with an alkyl
group (e.g., a methyl
group) or a phenyl group, to yield an ester. When the ester is administered to
a subject, the
ester is cleaved, enzymatically or non-enzymatically, reductively or
hydrolytically, to reveal
the anionic group. Such an ester can be cyclic, e.g., a cyclic sulfate or
sulfone, or two or
more anionic moieties may be esterified through a linking group. An anionic
group can be
esterified with moieties (e.g., acyloxymethyl esters) which are cleaved to
reveal an
intermediate TGR5 modulating compound which subsequently decomposes to yield
the
- 26 -
õõõ
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
active TGR5 modulating compound. In one embodiment, the prodrug is a reduced
form of a
carboxylate, sulfate or sulfonate, e.g., an alcohol or thiol, which is
oxidized in vivo to the
TGR5 modulating compound. Furthermore, an anionic moiety can be esterified to
a group
which is actively transported in vivo, or which is selectively taken up by
target organs.
The term "TGR5 modulator" means any compound that interacts with the TGR5
receptor. The interaction is not limited to a compound acting as an
antagonist, agonist, partial
agonist, or inverse agonist of the TGR5 receptor. In one aspect, the compounds
of the
present invention act as an antagonist of the TGR5 receptor. In another
aspect, the
compounds of the present invention act as an agonist of the TGR5 receptor. In
another
aspect, the compounds of the present invention act as a partial agonist of the
TGR5 receptor.
In another aspect, the compounds of the present invention act as an inverse
agonist of the
TGR5 receptor. The profile of a ligand, traditionally, endogenous or
synthetic, is
characterized by its intrinsic efficacy 'e' originally described by Furchgott
in 1966. It is used
to express the degree to which the different ligands produce varying
biological responses
while occupying the same number of receptors. Generally, the term "agonist"
means a
compound that enhances the activity of another molecule or receptor site. An
agonist, by
classical definition, whether a orthosteric, allosteric, inverse or a co-
agonist has a property to
bind to the receptor, alter its receptor state and result in a biological
action. Consequently,
agonism is defined as a property of an agonist or a ligand to produce a
biological action. In
contrast to this, an "antagonist" is essentially an agonist with high affinity
to the same
receptor macromolecule, but with very less or negligible intrinsic efficacy,
and thus sterically
prevents the biological actions of an agonist. As a property, antagonism may
be functional or
physiological, where an agonist has a direct competition for the receptor site
in former and
opposing effects via a different receptor-messenger system in the later. More
specifically, a
TGR5 agonist is a receptor ligand or compound that binds to TGR5 and increases
the
concentration of cyclic adenosine monophosphate (cAMP) by at least 20% in
cells expressing
the receptor." Conversely, a TGR5 antagonist would be a compound that
antagonizes or
blocks the activity of an agonist, thereby effecting a reduction in the
concentration of cAMP
The present invention relates to compounds having TGR5 receptor modulating
activity and their use to treat and prevent metabolic diseases such as
metabolic,
inflammatory, liver, autoimmune, cardiac, kidney, cancer and gasterintestinal
disease.
- 27 -
_
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Some representative compounds of the invention are shown below.
Compound Structure
No.
2A
OS03-Na+
HO'µ. 0H
H
3A
OH '=
S03-Na+
HO"4L.
H
4A
IMO SO3H
HO"
H z
6A OH
ire SO3H
7A OH "=-
e. SO3H
..01110
Ha
H OH
8A
0-S-0-H
API 2 8
H0' "OH
H
- 28 -
CA 02732323 2015-12-10
55929-4
9A
c61:c+\
QH -= 0
2 0-, =0,
H
"1-10''' . = 'OH
H i
\
Citation of publications and patent documents is not
intended as an admission that any is pertinent prior art, nor does it
constitute any admission
as to the contents or date of the same. The invention having now been
described by way of
written description, those of skill in the art will recognize that the
invention can be practiced
in a variety of embodiments and that the foregoing description and examples
below are for
purposes of illustration and not limitation of the claims that follow.
EXAMPLE 1: Synthesis of TGR5 Modulators
The compounds of the invention, and related derivatives, can be synthesized by
methods known to one skilled in the art.
- 29 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Example 1A: Synthesis of 3a,7a,12a-trihydroxy-6a-ethy1-513-cho1an-24-sulfonate
sodium
salt (3A)
õ 0 õ 0
OA: '.=
_
01 .
Pe OH OH
'OS c
a AcOss...''OR b
HO's
'Ac0".H :. = 'OR
____... H : --.-
1 2,R=Ac 3,R=Ac
2a,R=H 3a,R=H
OAc ''= c aril
Ac0". . OR Br
d, e ,
HO' . 'OH S03-Na
-\ -\
4,R=Ac 3A
4a, R=H
Reagents and condition: a) Et3N, 4-Pyrrolidin-Pyridine, Ac20, CH2C12, 0 C,
(2:2a = 70:30);
b) Et3N, C1COOEt, THF, NaBH4, 41% from 1 (3:3a = 70:30); c) Ph3P, Br2,
Imidazole,
CH2C12, 45% (4:4a = 70:30); d) Na2S03, Et0H; e) NaOH 5% in 1120, 93%. Overall
yield:
17%.
3a,7a,12a-triacetoxy-6a-ethyl-5p-cho1an-24-oic acid (2):
To a cooled (0 C) and stirred suspension of 1 (250 mg, 0.572 mmol) in
distilled
CH2C12 (5 ml) in presence of 4-Pyrrolidin Piridine (9 mg, 0.057 mmol),
distilled Et3N (0.78
ml, 5.430 mmol) was added was added. Subsequently acetic anhydride (0.49 ml,
5.140
mmol) was added dropwise under nitrogen atmosphere. After 20' the solution was
warmed to
room temperature and stirred overnight. The mixture was concentrated under
reduced
pressure, and the resulting residue was acidified with HC13N and extracted
with AcOEt
(3x15 ml). The organic layer was washed with brine (15 ml), dried over
anhydrous Na2SO4,
and concentrated under reduced pressure, to give 290 mg of mixture of 2 (70%)
and 3a,7a-
triacetoxy-7a-hydroxy-6a-ethyl-50-cholan-24-oic acid (2a, 30%) as resulted
from 11-1-NMR
analysis. The mixture was used for the following step without further
purification.
1H4s4MR (200MHz, CDC13) 5: 0.69 (3H, s, 18-CH3 of 2), 0.75 (3H, s, 18-CH3 of
2a), 0.80-
0.88 (m, 9H, m, 19-CH3 + 21-CH3 + CH2CH3 of 2 and 2a), 2.05 (9H, m, 3 x CH3C00-
of 2
-30-
,
4
1
1 _ õ-------õ-- --,-,-,õ ,---,- ,,,,,,,,,---,,
,,,,--,-_,-,,-õ,-- ,-,---,--,,--,-._ ------,------
1
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
and of 2a), 3.67 (1H, m. 7-CH-OH of 2a), 4.48 (1H, m, 3-CHOAc of 2 and of 2a),
5.01 (111,
m, 12- CHOAc of 2 and of 2a), 5.12 (114, m, 7-CHOAc of 2).
3a,7a,12a-triacetoxy -6a-ethy1-24-hydroxy-513-cho1ane (3):
To a solution of 2 (290 mg, crude of previous step) in distilled THE (35 ml)
under N2
atmosphere, distilled Et3N (0.75 ml, 5.148 mmol) and then ethylchloroformiate
(0.44 ml,
4.576 mmol) were added (NOTE: formation of white precipitate). Monitoring the
reaction by
TLC (petroleum ether/AcOEt 6:4) the mixture was stirred for 2 h until the
starting material
was completely reacted. To the mixture was then added dropwise NaBH4 (326 mg,
8.580
mmol) dissolved in H20 (3 ml), and the reaction mixture was stirred overnight.
H20 (30 ml)
and HC1 3N (10 ml) were added, and the mixture was extracted with AcOEt (3x20
ml). The
organic layer was washed with brine (15 ml), dried over anhydrous Na2SO4, and
concentrated
under reduced pressure. The resulting residue was purified by flash
chromatography on
BiotageTM (column: 12+M, from 5% to 55% of AcOEt in petroleum ether) to give
130 mg
(-41% from 1) of mixture of 3 (70%) and 3a, 1 2a-triacetoxy-7a,24-dihydroxy-6a-
ethyl-50 -
cholane (3a, 30%) as resulted from 11-1-NMR analysis. The mixture was used for
the
following step without further purification.
1H-NMR (200MHz, CDC13) 8: 0.66 (3H, m, 18-CH3 of 3 and of 3a), 0.75-0.85 (m,
911, m,
19- CH3 + 21-CH3 + CH2CH3 of 3 and of 3a), 3.55 (2H, m, 24-CH2OH of 3 and of
3a), 3.67
(m, .1H, . 7-CH-OH of 3a), 4.52 (m, 3-CHOAc of 3 and of 3a), 4.99-5.06 (2H, m,
12-
CHOAc of 3 and 3a and 7-CHOAc of 3).
3a,7a,12a-trihydroxy-6a-ethy1-513-cho1an-24-so1fonate (3A):
To a solution of triphenylphosphine (270 mg, crude of previous step) in
distilled
CH2C12 (4 ml) under N2 atmosphere, bromine (0.02 ml, 0.426 mmol) and imidazole
(0.02 ml,
0.426 mmol) were added, and the mixture was stirred for 10'. 3 (130 mg, 0.237
mmol)
diluted in distilled CH2C12 (4 ml) was then added dropwise, and the mixture
was stirred until
the starting material was completely reacted. CH2C12 (20 ml) was then added,
and the
reaction mixture was washed with H20 (3x20 ml), brine (15 ml), dried over
anhydrous
Na2SO4, and concentrated under reduced pressure. The resulting residue was
purified by flash
chromatography on BiotageTM (column: 12+M, from 5% to 55% of AcOEt in
petroleum
ether) to give 65 mg (-45%) as mixture of 4 (70%) and 3a, 1 2a-triacetoxy-
7a,24-dihydroxy-
6a-ethy-5P -cholane (4a 30%).
-31 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
This mixture was then solubilized in Et0H (10 ml), Na2S03 5% in H20 (9 ml) was
added dropwise, and the reaction was stirred at 90 for 12 h. The mixture was
the warmed at
room temperature and stirred overnight. The reaction mixture was then
concentrated under
reduced pressure, and the resulting residue was diluted with 1120 (15 ml),
acidified with HC1
3N and extracted with CH2C12 (3x15 ml). The organic layer was washed with
brine (15 ml),
dried over anhydrous Na2SO4, and concentrated under reduced pressure. The
resulting
residue was then treated with NaOH 5% in H20 (15 ml) and refluxed for 24 h.
The mixture
was concentrated under reduced pressure and the residue was diluted with H20
(15 ml),
washed with C112C12 (2x15 ml), acidified with HC13N and extracted with
AcOEt/Me0H 8:2
(3x25 m1). The organic layer was dried over anhydrous Na2SO4 and concentrated
under
reduced pressure. The resulting residue was purified by medium pressure
chromatography
(column: "RP-1 8 Lobar B", Me0H/H20 6:4, 50 psi) to give 3a,7a,12a-trihydroxy-
6a-ethyl-
5f3-cholan-24-sulfonate sodium salt, 3A (47 mg, 93%).
Mp: >280 C
111-NMR (400MHz, CD30D) 8: 0.72 (3H, s, 18-CH3), 0.90 (6H, m, 19-CH3 and
CH3CH2), 1.04 (3H, d, J = 6.51 Hz, 21-CH3), 1.89 (111, m, 23-CH), 2.70 (211,
m, 24-
CH2S03), 3.30 (111, m, 3-CH), 3.68 (1H, m, 7-CH), 3.98 (1H, m, 12-CH).
13C-MR (100.3MHz, CD30D) 8: 10.41, 11.41, 16.27, 21.20, 21.84
(2),22.54,26.57,27.14, 28.03, 29.38, 32.71, 34.59, 34.63, 35.05, 35.40, 40.06,
41.40,
41.48,45.25,45.85,46.63,
51.50, 69.58, 71.50, 72.40.
Example 1B: Synthesis of 3a,7a,23-trihydroxy-6a-ethyl-23(5)-methyl-24-nor-5p-
cholan-
23-sulfate sodium salt (2A)
-32-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
OH
OR
Si* HO' H 40
.0H0.
H
'RO"' OH
OH
3
a FR( = 11 e d(.g:VFAlc
õõ.
0
113-0H
,
OSO3Na
g, h
MO" '''OH Ac0",C ''5OH H 00õ ' H
H H H
===, 2A 7\
6 7
Reagent and conditions: a) Me0H, pTSA, us, 30 C, 2h, 97%. b) PhMgBr, THF,
reflux, 12h.
c) Et0H, HC1, reflux, 12h, 84% from 2. d) Ac20, pyridine, DMAP, THF, 25 C,
12h, 79%. e)
03, Me2S, CH2C12, Me0H, -78 C, 20 min, 70%. f) NaBH4, Me0H, TIE, 25 C, 12h,
48%
(S). g) PyS03, pfridine. h) 5% NaOH in Me0H, 43%. Overall Yield: 9%.
Methyl 3a,7a-dihydroxy-23-methy1-6a-ethy1-513-eho1an-24-oate (2)
To a solution of 1 (2.0 g, 4.6 mmol) in methanol (150 ml)p-TSA (0.2 g, 1.05
mmol)
was added and the mixture was treated under ultrasound for 90'. The solvent
was evaporated
under reduce pressure, the residue was dissolved in CHC13 (200 ml) washed with
a saturated
aqueous solution of sodium bicarbonate (2x100 ml), water (100 ml) and brine
(100 m1).The
organic layer was dried on anhydrous sodium sulfate and evaporated to dryness
to afford the
methyl ester 2 (2.0 g, 97%) as white solid.
3a,7a-cliihydroxy-23-methyl-6a-ethyl-513-bisnorcholanyldiphenylethylene(4)
To a solution of the methyl ester 2 (1.35g, 3.01mmol) in freshly distilled THF
(15 ml)
phenylmagnesiumbromide (24.1 ml, 1M in THF) was added and the resulting
mixture was
refluxed overnight. The solution was allowed to room temperature, a 3N
hydrochloric
solution (30 ml) was added dropwise and mixture was stirred for 30 min. The
organic phase
was separated and the aqueous one was extracted with Et0Ac (3x50 ml). The
combined
organic layers were washed with brine (1x100 ml), dried over anhydrous sodium
sulfate and
evaporated to dryness under reduced pressure, to get the intermediate 3, that
was used for the
following step without purification. The brown crude was dissolved in a
mixture of ethanol
(35 ml) and 12N HC1 (1 ml) and the mixture was refluxed overnight. The solvent
was then
- 33 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
evaporated in vacuo, the residue was dissolved in CH2C12 (100m1) washed with a
saturated
solution of sodium bicarbonate (2x60 ml), water (60 ml), brine (60 ml), dried
over anhydrous
sodium sulfate and evaporated under reduced pressure. The residue was purified
by flash
chromatography eluting with CHC13/Me0H 98:2 to afford the desired derivative 4
as white
solid (1.40 g, 84% from 2).
1H-NMR (CDC13) 8: 0.7 (3H, s, 18-CH3), 0.85 (3H, d, J= 6.5 Hz, 21-CH3), 0.88-
0.92
(6H, m, 19-CH3 + CH2CH3), 0.97-1.22 (611, m), 1.23-1.52 (9H, m), 1.57-1.70
(511, m), 1.78-
1.85 (4H, m), 1.92-1.95 (1H, m), 3.40-3.42 (111, m, 3-CH), 3.69 (1H, bs, 7-
CH), 7.25-7.29
(411, m, ArH), 7.127.23 (6H, m, ArH).
13C-NMR (CDC13) 8: 11.6, 11.7,
17.8,20.0,20.6,22.2,23.1,23.6,28.4,30.5,33.12,33.8,
34.8, 35.4 (2x), 39.4, 39.9, 41.1,
41.6,42.8,45.1,50.4,56.8,70.8,72.3,125.8,127.8(4x), 128.6
(4x), 129.8 (2x), 138.8, 143.4.
3a-acetoxy-7a-hydroxy-23-methy1-6a-ethy1-5P-bisnorcholanyldiphenylethylene(5).
To a solution of 4 (1.85g, 3.33 mmol) in freshly distilled THF (15 ml) acetic
anhydride (0.35 ml, 3.66 mmol), pyridine (0.05 ml, 0.66 mmol), 4-
di(methylamino)-pyridine
( 28 mg. 0.23 mmol) were added and the resulting mixture was stirred at room
temperature
overnight. The mixture was diluted with water (70 ml) and extracted with Et0Ac
(3x50 m1).
The combined organic layers were washed with water (2x50 ml), brine (50 ml),
dried over
anhydrous sodium sulfate and evaporated to dryness under reduced pressure to
get the desired
acetylated compound 5 (1 .57g, 79%) as white solid that was used for the next
step without
further purification.
1H-NMR (CDC13) 8: 0.65 (3H, s, 18-CH3), 0.85 (3H, d, J= 6.5 Hz, 21-CH3), 0.87-
0.92 (611, m, 19-CH3 + CH2CH3), 0.95-1.26 (8H, m), 1.30-1.50 (6H, m), 1.52-
1.70 (511, m),
1.75 (3H, s, 24- CH3), 1.79-1.97 (511, m), 2.01 (3H, s, 3-CHOC(0)CH3), 2.14-
2.17 (1H, m),
3.69 (111, bs, 7- CH), 4.52-4.57 (111, m, 3-CH), 7.11-7.22 (611, m, ArH), 7.25-
7.29 (4H, m,
ArH).
13C-NMR (CDC13) 6: 11.6, 11.8, 17.9, 20.1, 20.7, 21.4, 22.2, 23.1, 23.7, 26.6,
28.4,
29.6, 33.1, 34.9, 35.2, 35.5, 39.5, 40.0, 41.2, 41.6, 42.9, 45.1, 50.4, 56.9,
70.7, 74.7, 125.8,
127.8 (4x), 129.5 (4x), 129.8 (2x), 134.0, 138.9, 143.5.
- 34-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
3a-acetoxy-7a-hydroxy-6a-ethy1-513-cho1an-23-one(6)
A solution of 5 (1.35 g, 2.26 mmol) in a mixture of dry Me0H (15 ml) and dry
CH2C12 (20 ml) kept at -78 C was ozonized until the deep blue colour was
persistent for 30
min. The ozonization was then interrupted and the blue deep solution was swept
with
nitrogen to free the dissolved ozone. When the reaction is no more blue Me2S
(0.55 ml, 7.55
mmol) was added and the solution was allowed to return to room temperature.
The solvent
was then removed under reduced pressure and the residue was purified by flash
chromatography eluting with from 2% to 15% of Et0Ac in petroleum ether, to
afford the
desired ketone 6 as white solid (0.7 g, 70%).
1H-NMR (CDC13) 8: 0.7 (311, s, 18-CH3), 0.85-0.9 (9H, m, 19-CH3 + 21-CH3 +
CH2CH3), 1.04-1.23 (7H, m), 1.29-1.46 (8H, m), 1.58-1.72 (4H, m), 1.78-1.97
(5H, m), 2.00
(311, s, 3- CHOC(0)CH3), 2.11 (1H, s, 24-CH3), 2.43-2.53 (111, m), 3.70 (1H,
bs, 7-CH),
4.48-4.59 (1H, m, 3-CH).
13C-NMR (CDC13) 8: 11.6, 11.8, 19.7, 20.7, 21.5, 22.1, 23.1, 23.7, 26.6, 28.4,
29.6,
30.6, 32.6, 33.1, 35.1, 35.5, 39.5, 39.9, 41.1, 42.8, 45.0, 50.5, 50.9, 56.0,
70.7, 74.7, 170.7,
209.3.
3a-acetoxy-7a,23(S)-dihydroxy-6a-ethy1-511-cho1ane(7).
To a stirred solution of ketone 6 (0.5 g, 1.12 mmol) in THF at 0 C NaBH4 (0.27
g,
7.17 mmol) was added in one portion and then Me0H (0.27 ml, 6.7 6mmol) was
added
dropwise. At the end of the addition the reaction was warmed to room
temperature and stirred
overnight. The solvent was then evaporated to dryness, the residue was
dissolved in CH202
and 3N HC1 and stirred for lh. The two phases were separated, the organic
layer was washed
with water, brine, dried over anhydrous sodium sulphate and evaporated to
dryness under
reduced pressure. The oily residue, which consisted of two components as
evidenced by TLC,
was purified by flash chromatography (petroleum ether/Et0Ac 7:3) to afford
firstly the 3a-
acetoxy-7a,23(S)- dihydroxy-6a-ethyl-5f3-cholane 7 (0.225 g, 48%), then the
more polar 3a-
acetoxy-7a,23(R)- dihydroxy-6a-ethyl-5f3-cholane (0.105 g, 23%) in a 2:1
ratio.
1H4fM1 (CDC13) 8: 0.7 (3H, s, 18-CH3), 0.88-0.91 (6H, m, 19-CH3 + CH2CH3),
0.97 (3H, d, .1= 6.4 Hz, 21-CH3), 1.04-1.08 (411, m,), 1.19 (311, d, J 6.1, 24-
CH3), 1.26-1.56
(1211, m), 1.53-1.71 (5H, m), 1.76-1.96 (4H, m), 2.0 (311, s, 3-CHOC(0)CH3),
3.72 (1H, bs,
7-CH), 3.79-3.95 (111, m, 23-CH), 4.51-4.59 (111, m, 3-CH).
- 35 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
13C-NMR (CDC13) 8: 11.6, 11.8, 18.5, 20.7, 21.5, 22.1, 23.1, 23.7, 24.8, 26.6,
28.4,
29.6, 32.6, 33.1, 35.1, 35.5, 39.6, 39.9, 41.1, 42.8, 45.0, 45.8, 50.5, 56.7,
65.1, 70.7, 74.7,
170.7.
6a-ethyl-23(S)methy1-3a,7a,23-trihydroxy-24-nor-513-cholane-23 sulfate sodium
salt
(2A).
To a suspension of sulfurtrioxide pyridine complex (0.1 g, 0.49 mmol) in dry
pyridine
(5 ml), the alcohol 7 (0.2g, 0.49 mmol) was added and the resulting mixture
was stirred at
room temperature under nitrogen atmosphere for 48h after which the solvent was
evaporated
under reduced pressure. The residue was dissolved in 5% NaOH in Me0H (2m1) and
stirred
at room temperature overnight. The solvent was evaporated under reduced
pressure, the
resulting solid was in a mixture of 1120/CH3OH (1:1) and purified by reverse
phase
chromatography (column RP-18 lobar B) using a mixture of CH3OH/H20 (from 5:5
to 8:2) as
mobile phase, to afford the desired sulphate, 3a,7a,23-trihydroxy-6a-ethy1-
23(5)-methy1-24-
nor-513-cholan-23-sulfate sodium salt, 2A (0.1 1g, 43%).
111-NIVER (CD30D) 8: 0.7 (3H, s,18-CH3), 0.89-0.94 (611, m, CH3-19, CH2CH3),
1.01
(31I, d, J= 6.4 Hz, 21-CH3), 1.09-1.29(411, m,), 1.32 (3H, d, J= 6.1, 24-CH3),
1.36-1.41 (511,
m), 1.47- 1.61 (611, m), 1.71-1.89 (9H, m), 2.02-2.05 (1H, m), 3.28-3.35 (11I,
m, 3-CH), 3.66
(11I, bs, 7- CH), 4.54-4.58 (111, m, 23-CH).
13C-NMR(CD30D) 8: 12.0, 12.3, 19.4, 21.9, 22.4, 23.5, 23.7, 24.6, 29.4, 31.3,
33.5,
34.4, 34.5, 36.6, 36.7, 41.1, 41.5, 43.1, 43.8, 45.5, 46.9, 51.7, 58.1, 71.2,
73.2, 74.8.
EXAMPLE 2: In vitro TGR5 and FXR Activity
The potency and efficacy of the compounds of the invention on TGR5 receptor
was
evaluated using in vitro assays. Table 1 shows that compounds of the invention
are potent
and selective TGR5 modulators. In some aspects of the invention, the compounds
are dual
FXR and TGR5 agonists.
-36-
_
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Table 1: Potency and Efficacy of Compounds of the Invention on FXR and TGR5
Receptors
_____________________________________________________ Transactivation
FRET-cAMP
Alphascreen FRET Assay on
TGR5
(cA.MP)
overexpressing
Assay
NCI-H716
HEH293 cells
Compound hFXR hTGR5 hTGR5
hTGR5
(Reference (CDCA. 8-25 pNi ) (.1 CA=4-8 Of)
(LCA-6 (IX A=0,3-5 11.1)
Sfamdarci) EC,o EC0(11M) EC50(1M)
EC50(1M)
2A 0,145 0.05 1,6 0,3 1,2
0,025
(LCA=6,2) (LCA=0,3)
3A 7 3 0,7 0,2 0,6
0,011
(LCA = 6,2) (LCA = 0,3)
Cmp Chemical Structure TGR5 TGR5
d no EC50 efficacy
6A JMC2008
OH 1.00 M 103
37
so3H
Hoo.ew
7A JMC2008 OH 5.02 109
41
so3H
Has.4116.1.'10H
For a description of an in vitro TGR5 receptor binding assay, See, e.g.,
Kawamata, J.
Biol. Chem 2003, Vol. 278 No. 11, p. 9435-9440). Activity on FXR was assayed
by
fluorescence resonance energy transfer (FRET) for recruitment of the SRC-1
peptide to
human FXR using a cell-free ELiSA. See, Blanchard et al. WO 00/37077.
Luciferase
activity was determined in CHO cells stably expressing hTGR5 or transiently
cotransfected
with a hTGR5 expression vector and a cAMP-responsive element (CRE)-driven
luciferase
- 37 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
reporter gene. Some of the compounds were further submitted to a luciferase
reporter assay
to score for their capacity to activate the nuclear bile acid receptor FXR.
The following materials and methods were used, for example, to determine the
data in
Table 1.
Plasmids
The NIH Mammalian Gene Collection clone MGC:40597 (also named
pCMVSPORT6/hTGR5 or pTGR5) and pcDNA3.1(+) were obtained from Invitrogen
(Carlsbad, CA). pCRE-Luc and pCMV[3 were obtained from Clontech (Palo Alto,
CA).
pCMX-hFXR and pCMX-mRXRa were kind gifts from Dr. David J. Mangelsdorf
(Howard Hughes Medical Institute, University of Texas Southwestern Medical
Center).
pEcREx7-Luc was a kind gift from Dr. Richard A. Heyman (X-ceptor Therapeutics,
CA).
Cell culture
Chinese hamster ovary (CHO) cells, NCI-H716 cells, Hep3B cells and COSI cells
were obtained from American Type Culture Collection (Manassas, VA). Cell
culture medium, serum and supplements were from Invitrogen or Sigma-Aldrich.
All
CHO cells were maintained in minimum essential medium a (a-MEM) supplemented
with
10%(v/v) fetal bovine serum (FBS) and 100 M nonessential amino acids (NEAA).
NCI-H716
cells were maintained in suspension in RPMI-1640 supplemented with 10%(v/v)
FBS, 10mM
HEPES and 1mM sodium pyruvate. Hep3B cells were maintained in Eagle's medium
supplemented with 10%(v/v) FBS and 100 M NEAA. COSI cells were maintained in
Dulbecco's modified Eagle's medium (DMEM) supplemented with 10%(v/v) FBS. All
cell
culture medium was supplemented with 100units/m1 penicillin and 100 g/m1
streptomycin
sulfate. Cells were grown at 37 C in an atmosphere of 5% CO2, passed every 2-
6 days and
freshly plated for each experiment.
Transient transfections
CHO cells were plated in 96-well plates at a density of 3.5x104cells/well,
cultured for
24h, and then transfected with 15Ong of human (h) TGR5 expression plasmid
(pCMVSPORT6/hTGR5) and 10Ong of cAMP-responsive element (CRE)-driven
luciferase
reporter plasmid (pCRE-Luc) in each well using Lipofectamine 2000 reagent
(Invitrogen)
according to the manufacturer's instructions. After 6h incubation, cells were
washed once with
phosphate-buffered saline (PBS) and medium was exchanged for DMEM containing
0.1%(w/v)
-38-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
bovine serum albumin (BSA). After incubation for another 18h, cells were
treated for 5h
with different concentrations of each compound in fresh DMEM containing
0.1%(w/v) BSA.
After treatment, the cells were lysed with 50 1 of lysis buffer (25mM Dis-C1
(pH7.6), 2mM
EDTA, 1mM dithiothreitol (DTT), 10%(v/v) glycerol and 1%(v/v) triton X-100) by
a freeze-
thaw cycle and subjected to luciferase assays as described below.
COSI cells were plated in 96-well plates at a density of 2.5 x104cells/well in
DMEM
supplemented with 10%(v/v) charcoal-stripped FBS, cultured for 24h, and then
transfected
with 25ng of hFXR expression plasmid (pCMX-hFXR), 25ng of mouse (m) retinoid X
receptor a (RXRa) expression plasmid (pCMX-mRXRa), 5Ong of reporter plasmid
(pEcREx7-Luc) and 5Ong of pCMV13 as internal control in each well, using the
Lipofectamine
2000 reagent. After 24h, cells were washed twice with PBS and treated with
different
concentrations of each compound in fresh DMEM supplemented with 10%(v/v)
charcoal-
stripped FBS for 24h. After treatment, the cells were lysed with 50 1 of lysis
buffer by a
freeze-thaw cycle and subjected to both luciferase and 0-galactosidase assays
as described below.
Normalized luciferase values were determined by dividing the luciferase
activity by the (3-
galactosidase activity.
Luciferase and fl-galactosidase assays
For luciferase assays, 20111 of cell lysate was mixed with 100111 of
luciferase reaction
buffer [235 M luciferine, 265 M ATP and 135 M coenzyme A (CoA)] and
luminescence
was determined with CentroXS3 LB960 (Berthold Technologies, Bad Wildbad,
Germany).
For P-galactosidase assays, 10 1 of cell lysate was mixed with 100 1 of Buffer
Z [60mM
Na2HPO4, 10mM KC1, 1mM MgSO4, 50mM13-mercaptoethanol and 0.75mg/mlo-
nitropheny113-D-galactopyranoside (ONPG)] and incubated at 37 C for 0.5-3h.
Reactions
were stopped by adding 50 1 of Stop buffer (1M Na2CO3) and the optical density
at 420nm
was determined.
Establishing CHO cells stably expressing human TGR5 (CHO-TGR5 cells)
CHO cells were transfected with 3.8 g of hTGR5 expression plasmid
(pCMVSPORT6/hTGR5), 3.8 g of CRE-driven luciferase reporter plasmid (pCRE-Luc)
and
0.4n of neomycin-resistant gene expression plasmid [pcDNA3.1(+)] using
Lipofectamine 2000.
The transfectants were selected with 400 g/m1 G418 sulfate and single clones
were grown in
- 39-
6
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
96-well plate, independently. TGR5-expressing CHO cell lines were screened by
LCA
treatments, followed by luciferase assays.
cAMP production analysis
NCI-H716 cells were plated in 96-well plates coated with 0.75mg/m1 Matrigel
(BD
Biosciences) according to manufacturer's instructions just prior to use, at a
density of
6x104cells/well in DMEM supplemented with 10%(v/v) FBS, 100units/m1 penicillin
and
100 g/m1 streptomycin sulfate, and cultured for 24h, which allowed cell
adhesion to the
bottom of the plate. CHO-TGR5 cells were plated in 96-well plates at a density
of 3.5 x
104cells/well in a-MEM supplemented with 10%(v/v) FBS, 100 M NEAA, 100
units/ml
penicillin and 1001,tg of streptomycin sulfate, and cultured for 24h. The
cells were washed
twice with PBS and medium was exchanged for cAMP assay medium [DMEM containing
0.1%(w/v) BSA and 0.5mM 3-isobuty1-1-methylxanthine (IBMX)]. After incubation
for 30
minutes at 37 C, the cells were treated with each compound in fresh cAMP
assay medium for
30 minutes. After treatment, medium was discarded and cAMP amounts were
determined
using cAMP-Screen kit (Applied Biosystems, Foster City, CA) according to
manufacturer's
instructions.
50% effective concentrations (EC50) and efficacy determination
Assays were performed in triplicate or quadruplicate for each condition. EC50
values
were determined by probit analysis. Efficacy was determined by calculating
percentages of
10 M LCA value for TGR5 agonist study and lOpM 6a-Et-CDCA value for FXR
agonist
study, respectively. After subtracting the average value of the basal (vehicle-
treated) condition,
values were applied to EC50 and/or efficacy determinations. Calculation of
average EC50
and comparison of the EC50 between different compounds were done after
transformation to
logarithms.
Statistical analysis
Statistical analysis was performed by Student's t-test and p<0.05 was
considered
statistically significant.
Example 3 Metabolic activities of compounds of the invention in a diet-induced
obesity
mouse model
- 40 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
The goal of the study is to define whether TGR5 agonists (oleanolic acid (OA)
or
compounddof the invention (for example, a "test compound")) correct the
development of
obesity and associated insulin-resistance in vivo. To test this possibility,
OA/ test compound
are administered via food administration for 16 weeks to male C57BL6J mice
that were
previously subjected for 10 weeks to a high fat diet.
II- Protocol
In a previous study, OA was observed as a selective TGR5 agonist that did not
cause
food aversion. Animals treated with a dose of 100 mg/kg/day of OA showed,
however, some
signs of toxicity, whereas a lower dose was well tolerated. Therefore, OA is
administered at
the dose of 50 mg/kg/d in this study.
In vitro studies have identified compounds of the invention as potent and
selective
TGR5 ligands. No problems with toxicity are expected with compounds of the
invention,
which are administered at ¨ 50 ¨fold lower concentration.
For this study, 48 male C57BL6J mice (5 weeks of age) are divided in two
groups:
one group of 24 (group 1, 2&3) animals receives chow diet whereas the other
group of 24
receives a high fat diet for a period of 10 weeks (group 4,5&6). The animals
are then
analyzed during a period of 16 weeks. Five groups of 10 animals are assigned
as follows:
1: chow diet
2: chow diet + OA 50 mg/kg/day
3: chow diet + test compound e.g., 30 mg/kg/day
4: high fat diet
5: high fat diet + OA 50 mg/kg/day
6: high fat diet + test compound e.g., 30 mg/kg/day
During the entire study, body weight and food intake are monitored twice
weekly.
Week-2: Body composition is analyzed, for all groups, by dual energy X-ray
absorptiometry (dexascan).
Week-1: Serum levels of transaminases, glucose, triglycerides, cholesterol,
HDL-C,
LDL-C and insulin are measured in all groups after a fasting period of 12 h
and mice are then
placed on the diets as indicated (Day 0).
- 41 -
,
-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Week 2: Serum levels of transaminases, glucose, triglycerides, cholesterol,
HDL-C,
LDL-C and insulin are measured in all groups after a fasting period of 12 h
(Day 14).
Week 4: Glucose tolerance is determined by subjecting all the animals to an
intraperitoneal glucose tolerance test (IPGTT). Animals are fasted for 12 h
prior to this test.
Nocturnal energy expenditure of groups 1, 4, 5 and 6 (chow diet, high fat diet
and high fat
diet OA / test compound are measured by indirect calorimetry.
Week 8: Body weight composition is again analyzed by dexascan for all groups.
Serum levels of transaminases, glucose, triglycerides, cholesterol, HDL-C, LDL-
C and
insulin are measured in all groups after a fasting period of 12 h (Day 56).
Week 9: Circadian activity of groups 4, 5 and 6 (high fat diet fed mice) is
studied
during a period of 30 h.
Week 10: Measurement of blood pressure and heart rate is performed on groups
4, 5
and 6.
Week 11: Rectal temperature of all animals is measured at room temperature at
10:00
am.
Circadian activity measurement is performed on groups 1, 2, 3 and 4.
Week 12: Glucose tolerance is analyzed by performing an intraperitoneal
glucose
tolerance test (IPGTT) on groups 4, 5 and 6. During the IPGTT, blood is also
collected to
analyze insulin levels. Animals are fasted 12 h prior to these tests.
Feces are collected in all groups over a 24 h time period and fecal lipids
content is measured.
Week 16: Cold test is performed on all animals by measuring body temperature
of
animals exposed to 4 C.
Three days later, animals are sacrified. At sacrifice, blood is collected and
analyzed
for: plasma lipids (TC, TG, HDL-C, FFAs); liver functions (ALAT, ASAT,
alkaline Pase, 7-
GT); glucose and insulin; lipoprotein profiles of selected groups of plasma
(size-exclusion
chomatography).
Liver, small intestine, adipose tissues (WAT and BAT), pancreas, heart and
muscle
are collected, weighed and kept for further analyses including: standard
histology (HE
staining, succinate dehydrogenase staining, oil-red-0 staining and cell
morphology); tissue
lipid content; electron microscopy on BAT and muscle to analyze mitochondria;
RNA
isolation for expression studies of selected genes involved in metabolism and
energy
homeostasis by quantitative RT-PCR; Protein extraction for the study of post-
translationnal
modifications such as acetylation of proteins of interest (e.g. PGC-1a).
-42-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
III- Detailed procedures
A- Animal procedure and diets
Animals housing and handling
Mice are group housed (5 animals / cage) in specific pathogen-free conditions
with a 12 h:12
h (on at 7:00) light-dark cycle, in a temperature (20-22 c) and humidity
controlled vivarium,
according to the European Community specifications. Animals are allowed free
access to
water and food.
Drinking water
Chemical composition of the tap water is regularly analyzed to verify the
absence of potential
toxic substances at the Institut d'Hydrologie, ULP, Strasbourg. Drinking water
is treated with
HC1 and HC104 to maintain pH between 5 and 5,5 and chlorin concentration
between 5 and 6
PPIn=
Diet
The standard rodent chow diet is obtained from UAR and the high fat diet is
obtained from
Research Diet. Mice are fed, either with chow diet (16% protein, 3% fat, 5%
fiber, 5% ash) or
with high fat diet (20% protein, 20% carbohydrate, 60% fat). Oleanolic acid
and test
compound were mixed with either powdered chow diet or either powdered high fat
diet in the
following proportions: 0,5g of 0A/kg of food for the 50mg/kg/day treatment and
0,08g of test
compound /kg of food for the 10 mg/kg/day treatment. Pellets are then
reconstituted. Control
groups receive food pellets without test compound or OA. Due to the
consistency of the high
fat diet, no water is added in the mix with OA. In the case of the chow diet,
which is harder to
reconstitute, a minimal amount of water is added to the powder to reconstitute
pellets, which
are then air-dried. New batches of food are prepared weekly.
Blood collection
Blood is collected either from the retro-orbital sinus under anesthesia or
from the tail vein.
Anesthesia
For the dexa scanning experiment, animals are anesthesized with a mixture of
ketamine (200
mg/kg) / Xylasine (10 mg/kg) administred by intra-peritoneal injection.
For the venipuncture, animals are anesthesized by inhalation of an isofiurane-
02mixture.
B-Biochemistry
-43 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
The tests are performed with an Olympus AU-400 automated laboratory work
station using
commercial reagents (Olympus).
Analysis of lipids and lipoproteins
Serum triglycerides, total and HDL cholesterol are determined by enzymatic
assays. Serum
HDL cholesterol content are determined after precipitation of apo B-containing
lipoproteins
with phosphotungstic acid/Mg (e.g., Roche Diagnostics, Mannheim, Germany).
Free fatty
acids level are determined with a kit from Wako (e.g., Neuss, Germany) as
specified by the
provider.
Metabolic and endocrine exploration
Blood glucose concentration is measured by a Precision Q.I.D analyzer (e.g.,
Medisense
system), using Medisense Precis electrodes (e.g., Abbot Laboratories,
Medisense products,
Bedford, USA). This method is validated, by comparing Precision Q.I.D analyzer
values with
classical glucose measurements. The Precision Q.I.D method was chosen since it
requires a
minimal amount of blood and can hence be employed for multiple measurements
such as
during an IPGTT. Plasma insulin (e.g., Mercodia, Uppsala, Sweden) is
determined by ELISA
according to the manufacturer's specifications.
C-Metabolic testing
Lipoprotein profiles
Lipoprotein profiles are obtained by fast protein liquid chromatography,
allowing
separation of the three major lipoprotein classes VLDL, LDL, and HDL.
Intraperitoneal glucose tolerance test (IPGTT) - Oral glucose tolerance test
IPGTT is performed in mice which are fasted overnight (12 h). Mice are either
injected
intraperitoneally (IPGTT) with a solution of 20 % glucose in sterile saline
(0.9% NaCl) at a
dose of 2g glucose/kg body weight. Blood is collected from the tail vein, for
glucose and
insulin monitoring, prior and 15, 30, 45, 75, 90, 120, 150, 180 min after
administration of the
glucose solution. The incremental area of the glucose curve is calculated as a
measure of
insulin sensitivity, whereas the corresponding insulin levels indicate insulin
secretory
reserves.
Energy expenditure
Energy expenditure is evaluated through indirect calorimetry by measuring
oxygen
consumption with the Oxymax apparatus (e.g., Columbus Instruments, Columbus,
OH)
during 12 h. This system consists of an open circuit with air coming in and
out of plastic
- 44 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
cages (one mouse per cage). Animals are allowed free access to food and water.
A very
precise CO2 and 02 sensor measures the difference in 02 and CO2 concentrations
in both air
volumes, which gives the amount of oxygen consumed in a period of time given
that the air
flow of air coming in the cage is constant. The data coming out of the
apparatus is processed
in a connected computer, analyzed, and shown in an exportable Excel file. The
values are
expressed as ml.kg-1.1-11, which is commonly known as the V02.
Determination of body fat content by Dexa scanning
The Dexa analyses are performed by the ultra high resolution PIXIMUS Series
Densitometer
(0.18 x 0.18 mm pixels, GE Medical Systems, Madison, WI, USA). Bone mineral
density
(BMD in g/cm2) and body composition are determined by using the PIXIMUS
software
(version 1.4x, GE Medical Systems).
D-Non-invasive blood pressure measurement and pulse
The Visitech BP-2000 Blood Pressure Analysis System is a computer-automated
tail'cuff system that is used for taking multiple measurements on 4 awake mice
simultaneously without operator intervention. The mice are contained in
individual dark
chambers on a heated platform with their tails threaded through a tail cuff.
The system
measures blood pressure by determining the cuff pressure at which the blood
flow to the tail
is eliminated. A photoelectric sensor detects the specimen's pulse. The system
generates
results that have been shown to correspond closely with mean intra-arterial
pressure
measured simultaneously in the carotid artery. This allows reproducible values
of systolic
blood pressure and heart beat rate to be obtained. This requires training of
the animals for one
week in the system.
E- Circadian activity
Spontaneous locomotor activity is measured using individual boxes, each
composed
with a sliding floor, a detachable cage, and equipped with infra-red captors
allowing
measurement of ambulatory locomotor activity and rears. Boxes are linked to a
computer
using an electronic interface (e.g., Imetronic, Pessac, France). Mice are
tested for 32 hours in
order to measure habituation to the apparatus as well as nocturnal and diurnal
activities. The
quantity of water consumed is measured during the test period using an
automated
lickometer.
-45-
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Example 4. Physico-chemical properties
Water Solubility
Solid compound (BA) was suspended in 5 ml of 0.1 M HC1. The saturated
solutions,
after incubation for 1 week, were filtered on a Millipore filter (0.22 pm)
and the
concentration of BA is measured by HPLC-ESI-MS/MS using C18 column (150mm x
2mm
i.d., 4 m) and mobile phases of water containing 15mM acetic acid pH 5 and
acetonitrile.
The flow rate was 150 1/min. The mass spectrometry acquisition was performed
in the
multiple reaction monitoring mode using the ESI source in negative ionization.
Water
solubility was expressed as maliter.
The water solubility was measured for the insoluble protonated species of
carboxylated bile acids at a pH 1. Sulphate and sulphonate compounds 3A and 2A
are
ionized even at low pH and in physiological conditions are always soluble in
all biological
fluids.
Table 2: Physicochemical properties of the studied analogues and natural
occurring BA.
Bile Acid Ws(a) CMC(b) STcmc(c) LogPA_
Albumin
(d)
(111V1) 0,15 M Dyne/cm Binding (e)
Na+
(%)
(m1V1)
CDCA 32 3.2 45.5 2.2 93
UDCA 7.5 6.0 50.5 2.2 94
CA 273* 11* 1.1* 50*
TCDCA hs 3.0* 0.9* 70*
TUDCA hs 2.2* 1.1* 67*
6MUDCA 28* 4.2* 1.3* 80*
3A hs 2.8 43.4 0.7 81
2A hs Not available Not available 2.3 97
a Ws: water solubility refers to BA as protonated species and therefore not
evaluated for 3A, 2A,
TCDCA, and TUDCA which are highly soluble (hs).
b CMC: Critical Micellar Concentration determined in 0,15 M NaC1 water
solution.
-46-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
c ST cmc: Surface Tension at CMC in 0,15 M NaC1 water solution.
d LogPA: 1-octanol-water partition coefficient of the studied bile acids as
ionized species.
*: values from literature.
Critical Micellar Concentration (CMC)
The detergency i.e. the tendency to form micelles was evaluated for charged
molecules which are soluble in water as Sodium salt (2 unit up the pKa). The
critical
micellar concentration (CMC) was determined by surface tension (ST)
measurements using a
maximum bubble-pressure method which give surface tension values slightly
affected by
potential impurities like static methods are. The tensiometer was a Sensadyne
6000 (Chem-
Dyne Research Corp., Milwaukee, WI) equipped with two glass probes of 0.5 and
4.0 mm
diameters connected to a source of nitrogen. The bubble frequency was 1
bubble/second in
distilled water at 26 C (P=2.7 atm) and the calibration was made with double-
distilled water
and methanol. The surface tension of BA sodium salts solutions in NaCl 0.15 M
was
measured at various concentrations inside the 0.13-50 mM range. The surface
tension values
were plotted against the logarithm of the bile salt concentration; the
regression lines
corresponding to the two parts of the curve (monomeric and micellar phases)
were calculated
using the method of least squares, and the intersection of the lines was taken
as the CMC
value. From the ST vs concentration curves the value of the surface tension at
the CMC
(equilibrium between monomers and multimers species) was also calculated
giving
information about the detergency power which is related to the size of the
micelles with
associate surface tension lowering capacity.
The CMC was evaluated by surface tension measurements in non equilibrium
conditions i.e. in conditions that impurities slightly affect the surface
tension results (Figure
1). Compound 3A presents a high surface tension lowering capacity with a CMC
of 2.8 mM.
The behaviour of the ST vs concentration suggests the formation of relatively
large aggregate
with consistent detergency.
Octanol/water partition coefficient
Since the sulphate and sulphonated analogues are always ionised at all pH
values the
octanollwater partition coefficient was measured for all molecules in ionized
form. 1-
Octanol/water partition coefficient (log P) was evaluated using a conventional
shake-flask
- 47 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
procedure. The experiments were carried out on 0.1 mM bile salt solution
buffered at pH 8
with 0.1 M phosphate buffer to ensure complete ionization of the BA; the log P
values refer
to the BA in the ionized form, not to the protonated species, and the initial
concentration of
each BA was below its own CMC value. The aqueous buffer was previously pre-
saturated
with 1-octanol, 5 ml of 1-octanol pre-saturated with water was then added and
the samples
were left to equilibrate for 2 weeks under continuous stirring at room
temperature After
centrifugation the two phases were carefully separated. BA concentration in
the water phase
was measured with HPLC-ESI¨MS/MS using C18 column (150mm x 2mm i.d., 41.tm)
and, as
mobile phases, water containing 15 mM acetic acid pH 5 and acetonitrile. The
flow rate was
150 al/min and the column was maintained at 45 C. The mass spectrometry
acquisition was
performed in the multiple reaction monitoring mode using the ESI source in
negative
ionization.
The 1-octanol/water partition coefficient was calculated for the ionized
species to
facilitate the comparison between the carboxyl and sulphate bile acids since
the latter did not
protonate even at very low pH value.
Albumin binding
The extent of albumin binding was evaluated by equilibrium dialysis at a fixed
BA-
albumin ratio. BA was dissolved at a concentration of 100 1.1M in 5% bovine
serum albumin-
saline solution (pH 7.2) and left to stand for 24 h at 25 C. Two millilitres
of this solution was
dialyzed in cellulose sacs having a molecular weight cut-off of 12-14,000
against 25 ml of
saline solution. The system was equilibrated by mechanical gently shaking for
72 h at 25 C.
BA concentrations of the dialyzed solution (corresponding to the free unbound
fraction) and
of the starting solution were determined with HPLC-ESI-MS/MS in the same
conditions of
the previous analysis. The percent of albumin binding was calculate from the
initial BA
concentration and from the unbound concentration in the dialyzed fraction.
Data are reported
in the Table 2. All these compounds never less present an albumin binding
compatible with a
relatively fast hepatic uptake, similarly to natural occurring BA. Accordingly
different
kinetics (slower) of the liver uptake is expected.
Example 5. In vitro metabolic stability in human stool culture
Stability to Intestinal Bacteria.
7a-dehydroxylation.
-48-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Homogenized fresh human stools (500 mg) were transferred into sterile vials to
which
mL of sterilized chopped meat-glucose medium (Scott Lab., Fiskville, RI) was
added. BA
were then added at a final concentration of 0.05 mM. Vials were incubated at
37 C; then, at
0, 1, 2, 4, 8 and 24 h after the addition of the BA, the reaction was stopped
with 150 L, of
5 30% KOH. The samples were centrifuged at 3500 rpm for 10 min; from the
supernatant the
BA were isolated by C-18 solid-phase extraction and analyzed by TLC and HPLC-
ES-
MS/MS.
Thin-layer chromatography (TLC), utilizing silica gel 0.25 mm thickness plates
(Merck, Darmstat, Germany), was employed as the first screening test. The
solvent system
used for the separation of conjugated BA was composed of propionic
acid/isoamyl
acetate/water/N-propanol (3:4:1:2, v/v/v/v; solvent I), and that of the
unconjugated BA was
acetic acid/carbon tetrachloride/isopropyl ether/isoamyl acetate/water/N-
propanol/benzene
(1:4:6:8:2:2, v/v/v/v/v/v; solvent II). Separated BA were revealed with 5%
phosphomolybdic
acid ethanol solution.
All the studied analogues are very stable when incubated in human stool
cultures and
even after 24 hour more than 85% of the compounds can be recovered unmodified
as
reported in Fig 2. On the contrary the reference natural analogue CDCA
presents an half-life
time of almost one hour and after 8 hours of incubation is almost completely
metabolized (7-
dehydroxylated) to form lithocholic acid. After also long time incubation for
all 6 ethyl
analogues the 7 dehydroxylation and the intermediate formation of a 7 oxo
derivative is
practically abolished.
Side chain stability
According to the first results the side chain is not modified by the
intestinal bacteria
enzymatic activities. These data suggest that the presence of the ethyl group
in the 6 position
protects the 7 hydroxyl group toward oxidation or removal by steric hindrance.
In addition
all the analogues are very stable also for side chain metabolism. The side
chain ester bond of
sulphate analogue 2A is quite stable in the human stool culture. The
sulphonate analogue 3A
is also very stable. No minor metabolites have been found by HPLC-ES-MS/MS.
These data
suggest that in the lower intestinal content in presence of anaerobic bacteria
these analogues
are stable.
- 49 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Example 6. In vitro metabolic stability in simulated duodenal/pancreatic fluid
(USP
specification)
This study has been performed for 2A since it contains an ester bond in the
side chain
and the aim was to verify the stability in presence of esterase enzymes like
present in
duodenal and pancreatic juice. Simulated pancreatic fluid was prepared by
dissolving 10 g/L
Pancreatin (Sigma P8096: pancreafin from porcine pancreas, activity lx USP
specifications)
in 0.05M phosphate buffer, pH = 7.2 0.1. Then, 4-mL aliquots of the
simulated pancreatic
fluid were added of 50 pM of the studied BA and incubated for different times
(0, 30, 60, 90,
120, 180 and 240 min) at 37 C. After incubation, a 2-mL aliquot of each
solution was added
with 2 mL of 0.1M NaOH and subjected to bile acids extraction by SPE and
analysis by thin-
layer chromatography and mass spectrometry as described above. Figure 2 shows
the
metabolic stability of 2A in simulated pancreatic fluid. Olive oil was used as
a reference as
reported in the USP protocol. The compound is very stable (see Fig. 2) and the
ester bond
(sulphate) in 2A is not hydrolyzed by pancreatic esterases suggesting an high
stability in
human duodenal and upper intestine content.
Example 7. Binary secretion and metabolism of Compound 3A in bile-fistula rat
after=
duodenal (id) and femoral (iv) administration
Aim and Rationale
The structural modification of the new analogues could affect their hepatic
uptake,
hepatic transports and secretion and intestinal absorption. Therefore the
knowledge of the
biliary secretion after both iv and id administration together their
metabolism is a key point in
the candidate selection for additional studies.
To evaluate the mode and efficiency of the intestinal absorption the analogues
were
administered both intravenously (femoral infusion) and orally (duodenal
infusion) at the same
dose and their biliary secretion rate was evaluated in bile fistula rat model.
The differences in
the area under the curve of the biliary secretion vs time between iv and id
administration
account of its intestinal absorption and give information about its
biovailability.
Choleretic effect
-Duodenal infusion
-50-
1
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
The bile fistula rat model was developed at the University of Bologna Lab
facilities.
The compound 3A was administered at a dose of 1 gmol/kg/min (1 hour infusion)
to a rat
group via duodenal infusion (id). Rat have a bile fistula to collect bile
samples at different
times before and during the infusion. For duodenal infusion experiment 3 rats
(250 10 g)
were treated. Bile samples were collected every 15 minutes for four hours. In
addition, 3
control rats were treated with saline solution under the same conditions for
times and
sampling (duodenal control rats).
Figure 3 shows bile flow during sample collection (one animal). Duodenal
infusion
starts after 30 min baseline bile collection and continues for one hour.
Compound 3A
presents a moderate choleretic effect.
-Intravenous infusion
For femoral infusion experiment, 3 rats were treated. Figure 4 shows bile flow
during
the study. Femoral infusion starts after 75 minutes of steady-state and
continues for 60 min.
Bile samples were collected every 15 minutes for four hours. In addition, 3
rats were treated
with saline solution under the same conditions for times and sampling (femoral
control rats).
Compound 3A presents a moderate choleretic effect and similar to that obtained
in the
id experiment
Biliary secretion of the administered analogues
Bile samples collected during the iv and id experiments were analyzed to
determine
the biliary secretion of the administered analogues and their metabolites.
HPLC-ES-MS/MS analysis. Stock solutions of each compound in methanol at 1
mmol/L
(with the exception of UPF 1212 at 350 mon) were prepared and working
solutions were
prepared by diluting appropriate volumes of the primary solution. Methanol and
acetronitrile
- was of HPLC-grade purity. Ammonia was 30% and acetic acid was 99.8%. All
reagents
were obtained from Carlo Erba Reagents. HPLC-grade water was prepared by a
Milli-Q
system.
Sample preparation
Rat bile samples were brought to room temperature, briefly stirred, and
diluted 1:100
v/v (bile samples from duodenal orinfasion) and 1:100 or 1:200 v/v (bile
samples from
femoralr infusion) with 15 mM ammonium acetate buffer (pH =
5.0):acetonitrile=70:30 (v/v).
-51-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Final solution was transferred in autosampler vials, and 10 L was injected
into the
chromatographic column.
HPLC- ESI-MS/MS method
Bile rat samples were analyzed by liquid chromatography-tandem mass
spectrometry
(HPLC-MS/MS) using electrospray (ESI) source in negative ionization mode. For
liquid
chromatography a Waters Alliance 2695 separation module coupled with
autosampler was
used. Autosampler was maintained at 7 C. Separation was performed on a Synergi
Hydro-
RP C18 column (150x2.0mm i.d., 4 um particle size), protected by a
SecurityGuard ODS
4x2.0mm i.d. precolumn, both supplied from Phenomenex. Analyte was eluted
using 15 mM
ammonium acetate buffer (pH = 5.00) as mobile phase A and acetonitrile as
mobile phase B.
Mobile phase B was increased from 30% to 64% in 10 min, then to 100% in 10
min, and held
constant for 10 min. Flow rate was 150 L/min and the column was maintained at
45 C.
The column effluent was introduced into ESI source connected to a triple
quadruple mass
spectrometer (Quattro-LC, Micromass) operating in Multiple Reaction Monitoring
(MRM)
acquisition mode. Nitrogen was used as nebulizer gas at 90 L/h flow rate and
as desolvation
gas at 930 L/h. Ion source block and desolvation temperatures were set
respectively to 80 C
and 180 C. Capillary voltage was 3.0 kV. MassLynx software version 4.0 was
used for data
acquisition and processing. In addition, using mass spectrometry both in
single MS or
tandem MS/MS configuration experiments were performed to identify metabolites.
Quantification
A 5-point calibration curve was daily prepared and injected in duplicate.
Calibration
samples were obtained in the 0.1 to 25 mon concentration range prepared in
mobile phase.
Linear calibration curve parameters were obtained from the plot of the analyte
peak area
versus analyte concentration using a least squares regression analysis (weight
= 1/x2).
Correlation coefficients were >0.989.
Pharmacokinetic (biliary secretion) of the administered analogues: iv versus
id comparison
The data refer to the secretion rate of the analogues recovered in bile as
such after
duodenal and femoral infusion at a dose of lumoVKg/min. Major and minor
metabolites are
reported later. Table 3 shows compound 3A concentration and secretion values
obtained
from bile rat samples collected during the duodenal infusion (1 h ranging from
75 to 135
-52-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
min). Table 4 shows compound 3A concentration and secretion values obtained
from bile rat
samples collected during the femoral infusion (1 h ranging from 75 to 135 mm)
Table 3.
Time 3A
(min)
Conc. Secretion
(mmoUL) ( mol/kg/min)
90 n.d.b
2
120 0.37 0.026
150 0.90 0.079
180 1.3 0.101
210 0.95 0.061
240 0.70 0.043
270 0.58 0.039
300 0.51 0.029
a-: not calculated n.d.; not detected
Table 4.
Time 3A
(min)
Conc. Secretion
(mmol/L) ( mol/kg/min)
75 n.d.b
90 2.6 0.105
120 7.3 0.588
150 7.5 0.463
180 6.2 0.363
210 3.4 0.247
240 1 0.045
a-: not calculated n.d.; not detected
- 53
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
The biliary secretion of compound 3A (sulphonated analogue) after iv
administration
is very efficient and the compound is almost completely recovered in bile. The
kinetic profile
indicates that the analogue is efficiently taken up by the liver and secreted
in bile almost as
such without hepatic metabolism (Fig. 5). On the contrary after id
administration the
recovery in bile is much lower than the recovery after iv administration
suggesting that the
compound is poorly absorbed by the intestine (Fig. 5). According to the
physicochemical
properties, this compound is poorly absorbed by passive diffusion mechanism
(low LogP =
0.71) and active mechanism does not seem to be involved. The presence of three
hydroxyl
groups renders the molecule on one side efficiently taken up by the liver and
secreted into
bile but on the other side poorly absorbed by the intestine.
Example 8. Hepatic metabolism
For a preliminary screening the search of the possible metabolites has been
performed
on the basis of the expected compounds according to previous experiments and
data and to
the structure and physicochemical properties of the studied analogues.
Compound 3A is not metabolized by the liver or by the intestine and once taken
up by
the liver is secreted in bile unmodified with a fast kinetic resulting in a
low hepatic residence
time. When administered via id is poorly absorbed by the intestine and also in
this case is not
metabolized. Due to its relatively high hydrophylicity (low LogP) and to the
presence of the
sulphonate group and three hydroxyls the compound 3A is enough polar to be
secreted in bile
as such but too much polar to be absorbed by passive mechanism by the
intestine. Figure 6
shows compound 3A and its main metabolite identified in bile using mass
spectrometry in the
di experiment. Data are reported as absolute area values. Figure 7 shows a
zoom display of
Figure 6.
Example 9. Compound in vitro toxicity on HepG2 cell
Compounds of the invention were evaluated for cytotoxicity using standard
methods
known in the art. Specifically, cytotoxicity was evaluated using HEPG2 cells
and monitoring
ATP decrease and determining the EC50 value. Both compound 3A and 2A showed an
EC50
>1000 mM.
- 54 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Compound HepG2 cell Cytotoxicity
ATP decrease
EC50 ( ,M)
Staurosporine 15
Tamoxifen 47
LCA 84
CDCA 650
UDCA >1000
CA >1000
3A >1000
2A >1000
Example 10. NR Selectivity Assays
The selectivity of compounds of the invention was evaluated using assay
methods
known in the art. Specifically, the following assay methods were used:
FXR and TAR: Coactivator Recruitment (alphascreen);
TGR5: cAMP level on human intestinal cell line (NCI-H716);
PXR: Ligands Competition assay (Binding Assay)
CAR: Coactivator Recruitment (Lanthascreen)
Compound FXR TGR5 LXRa PXR CAR PPARE, VDR
(Ref (CDCA= (LCA = (T090131 (SR-12183= (CITCO= (0,004 M) (0,005W)
standard) 10-20 uM) 4-8 M) 7= 0,013[tM) 0.005 IIM)
EC50 EC50 (as.4)
EC50 (p.M) EC50 0,08 11.M) EC50 (.1M) EC50
(PM) EC50 (I-LM)
(I-LM)
CDCA 20 30 No >250 >250 No No
activity activity activity
LCA No 4-8 No 23 No No No
activity activity activity activity activity
CA No 30 No No No No No
activity activity activity activity activity activity
UDCA >150 No No >250 >250 No No
activity activity activity activity
2A 0.14 1.6 No 21 84 No No
activity activity activity
3A 7 0.7 No 150 >250 No No
activity I activity activity
,
-55-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
Example 11: Metabolic activity of compound 3A in a diet-induced obesity mouse
model
The goal of the study was to assess the therapeutic potency of compound 3A in
a
mouse model of diet-induced obesity. Compound 3A was administered via food
administration for 16 weeks to male C57BL6J mice that were previously
subjected for 10
weeks to a high fat diet.
II- Protocol
Compound 3A was administered for this study.
For this study, male C57BL6J mice (5 weeks of age) were divided in 2 groups:
one
group of animals received chow diet to control the onset of obesity, one group
of animals
received a high fat diet. Compound 3A was tested on the animals that received
the high fat
diet (HFD). The animals are then analyzed during a period of 13 weeks. The
groups were as
follows:
1: chow diet
2: high fat diet
3: high fat diet + 3A
During the entire study, body weight and food intake were monitored twice
weekly.
The period for the onset of obesity was 10 weeks. At the end of the 10 weeks,
body
composition was analyzed by dual energy X-ray absorptiometry (dexascan) and
serum levels
of transaminases, glucose, triglycerides, cholesterol, HDL-C, LDL-C and
insulin were
measured.
After 1 week: Compound was administered.
After 3 weeks: Blood collection. Serum levels of transaminases, glucose,
triglycerides, cholesterol, HDL-C, LDL-C and insulin were measured in all
groups after a
fasting period of 12 h.
After 2 weeks: Body weight composition was again analyzed by dexascan for all
groups.
After 2 weeks: Nocturnal energy expenditure was measured by indirect
calorimetry
and actimetry.
After 2 weeks: Glucose tolerance was determined by subjecting all the animals
to an
intraperitoneal glucose tolerance test (IPGTT). Animals were fasted for 12 h
prior to this test.
-56-
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
After 2 weeks: Animals were sacrified. At sacrifice, blood was collected and
analyzed for: plasma lipids (TC, TG, HDL-C, FFAs); liver functions (ALAT,
ASAT, alkaline
Pase, y-GT); glucose and insulin; lipoprotein profiles of selected groups of
plasma (size-
exclusion chomatography).
Liver, small intestine, adipose tissues (WAT and BAT), pancreas, heart and
muscle
were collected, weighed and kept for further analyses including: standard
histology (HE
staining, succinate dehydrogenase staining, oil-red-0 staining and cell
morphology); tissue
lipid content; electron microscopy on BAT and muscle to analyze mitochondria;
RNA
isolation for expression studies of selected genes involved in metabolism and
energy
homeostasis by quantitative RT-PCR; Protein extraction for the study of post-
translationnal
modifications such as acetylation of proteins of interest (e.g. PGC-1a).
III- Detailed procedures
A- Animal procedure and diets
Animals housing and handling
Mice were group housed (5 animals / cage) in specific pathogen-free conditions
with a 12
h:12 h (on at 7:00) light-dark cycle, in a temperature (20-22 c) and humidity
controlled
vivarium, according to the European Community specifications. Animals were
allowed free
access to water and food.
Drinking water
Chemical composition of the tap water was regularly analyzed to verify the
absence of
potential toxic substances at the Institut d'Hydrologie, ULP, Strasbourg.
Drinking water was
treated with HC1 and HC104 to maintain pH between 5 and 5,5 and chlorin
concentration
between 5 and 6 ppm.
Diet
The standard rodent chow diet was obtained from UAR and the high fat diet was
obtained
from Research Diet. Mice were fed, either with chow diet (16% protein, 3% fat,
5% fiber, 5%
ash) or with high fat diet (20% protein, 20% carbohydrate, 60% fat). Compound
3A was
mixed with powdered high fat diet. Pellets were then reconstituted. The HFD
control group
received food pellets without compound 3A. In the case of the chow diet, which
is harder to
reconstitute, a minimal amount of water was added to the powder to
reconstitute pellets,
which are then air-dried. New batches of food were prepared weekly.
Blood collection
- 57 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Blood was collected either from the retro-orbital sinus under anesthesia or
from the tail vein.
Anesthesia
For the dexa scanning experiment, animals were anesthesized with a mixture of
ketamine
(200 mg/kg) / Xylasine (10 mg/kg) administred by intra-peritoneal injection.
For the venipuncture, animals were anesthesized by inhalation of an isoflurane-
02mixture.
B-Biochemistry
The tests were performed with an Olympus AU-400 automated laboratory work
station using
commercial reagents (Olympus).
Analysis of lipids and lipoproteins
Serum triglycerides, total and HDL cholesterol were determined by enzymatic
assays. Serum
HDL cholesterol content were determined after precipitation of apo B-
containing lipoproteins
with phosphotungstic acid/Mg (e.g., Roche Diagnostics, Mannheim, Germany).
Free fatty
acids level were determined with a kit from Wako (e.g., Neuss, Germany) as
specified by the
provider.
Metabolic and endocrine exploration
Blood glucose concentration was measured by a Precision Q.I.D analyzer (e.g.,
Medisense
system), using Medisense Precis electrodes (e.g., Abbot Laboratories,
Medisense products,
Bedford, USA). This method was validated, by comparing Precision Q.I.D
analyzer values
with classical glucose measurements. The Precision Q.I.D method was chosen
since it
requires a minimal amount of blood and can hence be employed for multiple
measurements
such as during an IPGTT. Plasma insulin (e.g., Mercodia, Uppsala, Sweden) were
determined
by ELISA according to the manufacturer's specifications.
C-Metabolic testing
Lipoprotein profiles
Lipoprotein profiles were obtained by fast protein liquid chromatography,
allowing
separation of the three major lipoprotein classes VLDL, LDL, and HDL.
Intraperitoneal glucose tolerance test (IPGTT) - Oral glucose tolerance test
IPGTT was performed in mice which were fasted overnight (12 h). Mice were
either injected
intraperitoneally (IPGTT) with a solution of 20 % glucose in sterile saline
(0.9% NaCl) at a
dose of 2g glucose/kg body weight. Blood was collected from the tail vein, for
glucose and
insulin monitoring, prior and 15, 30, 45, 75, 90, 120, 150, 180 min after
administration of the
- 58 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
glucose solution. The incremental area of the glucose curve was calculated as
a measure of
insulin sensitivity, whereas the corresponding insulin levels indicate insulin
secretory
reserves.
Energy expenditure
Energy expenditure was evaluated through indirect calorimetry by measuring
oxygen
consumption with the Oxymax apparatus (e.g., Columbus Instruments, Columbus,
OH)
during 12 h. This system consists of an open circuit with air coming in and
out of plastic
cages (one mouse per cage). Animals were allowed free access to food and
water. A very
precise CO2 and 02 sensor measures the difference in 02 and CO2 concentrations
in both air
volumes, which gives the amount of oxygen consumed in a period of time given
that the air
flow of air coming in the cage is constant. The data coming out of the
apparatus was
processed in a connected computer, analyzed, and shown in an exportable Excel
file. The
values were expressed as ml.kg-1.111, which is commonly known as the V02.
Determination of body fat content by Dexa scanning
The Dexa analyses were performed by the ultra high resolution PIXIMUS Series
Densitometer (0.18 x 0.18 mm pixels, GE Medical Systems, Madison, WI, USA).
Bone
mineral density (BMD in g/cm2) and body composition were determined by using
the
PIXIMUS software (version 1.4x, GE Medical Systems).
D-Non-invasive blood pressure measurement and pulse
The Visitech BP-2000 Blood Pressure Analysis System is a computer-automated
tail'cuff system that is used for taking multiple measurements on 4 awake mice
simultaneously without operator intervention. The mice were contained in
individual dark
chambers on a heated platform with their tails threaded through a tail cuff.
The system
measures blood pressure by determining the cuff pressure at which the blood
flow to the tail
is eliminated. A photoelectric sensor detects the specimen's pulse. The system
generates
results that have been shown to correspond closely with mean intra-arterial
pressure
measured simultaneously in the carotid artery. This allows reproducible values
of systolic
blood pressure and heart beat rate to be obtained. This requires training of
the animals for one
week in the system.
E- Circadian activity
- 59 -
CA 02732323 2011-01-27
WO 2010/014836 PCT/US2009/052290
Spontaneous locomotor activity was measured using individual boxes, each
composed
with a sliding floor, a detachable cage, and equipped with infra-red captors
allowing
measurement of ambulatory locomotor activity and rears. Boxes were linked to a
computer
using an electronic interface (e.g., Imetronic, Pessac, France). Mice were
tested for 32 hours
in order to measure habituation to the apparatus as well as nocturnal and
diurnal activities.
The quantity of water consumed was measured during the test period using an
automated
lickometer.
The results of this study are shown in Figures 8, 9, and 10. Figure 8 shows
the
evolution of weigh gain after treatment (%). The graph shows % weight gain (0,
5, 10, 15,
20, 25) verses time (0-9 weeks). Body weight gain is blunted in the HF-fed
mice treated with
compound 3A. Figure 9 shows the evolution of body composition after treatment
(%) 5
weeks after treatment. Graphs with % weight gain are shown for fat, lean and
body fluid
measurements. Compound 3A reduces fat gain upon HFD. Figure 10 shows an
assessment
of glucose homeostasis for compound 3A. Figure 10A shows glycemia 3 weeks
after
treatment. Specifically, Figure 10A is a bar graph that compares 3 groups of
mice (x-axis):
mice on HFD administered compound 3A(first bar), mice on a HFD (second bar),
and mice
on a chow diet (third bar) and their fasting glycemia (y-axis) mmo1/1 at 10,
12, 14, 16, 18, 20,
and 22. Figure 10B shows fructosamines 3 weeks after treatment. Specifically,
Figure 10B
is a bar graph that compares 3 groups of mice (x-axis): mice on HFD
administered
compound 3A(first bar), mice on a HFD (second bar), and mice on a chow diet
(third bar)
and their fructosamines (y-axis) micromole at 170, 175, 180, 185, 195, 200,
205, and 210.
Figure 10C shows the results of a glucose tolerance test 9 weeks after
treatment.
Specifically, Figure 10C is a graph that compares 3 groups of mice: mice on
HFD
administered compound 3A, mice on a RFD, and mice on a chow diet. The y-axis
shows the
amount in mg/dL (0, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500) and the x-
axis shows
time in minutes 0, 30, 60, 90, 120, 150, and 180.
1
Other Embodiments
While the invention has been described in conjunction with the detailed
description
thereof, the foregoing description is intended to illustrate and not limit the
scope of the
invention, which is defined by the scope of the appended claims. Other
aspects, advantages,
and modifications are within the scope of the following claims. It will be
understood by
- 60 -
CA 02732323 2011-01-27
WO 2010/014836
PCT/US2009/052290
those skilled in the art that various changes in form and details may be made
therein without
departing from the scope of the invention encompassed by the appended claims.
- 61 -